Next Article in Journal
Tough Bioplastics from Babassu Oil-Based Acrylic Monomer, Hemicellulose Xylan, and Carnauba Wax
Next Article in Special Issue
The Exosome-like Vesicles of Giardia Assemblages A, B, and E Are Involved in the Delivering of Distinct Small RNA from Parasite to Parasite
Previous Article in Journal
Emerging Personalized Opportunities for Enhancing Translational Readthrough in Rare Genetic Diseases and Beyond
Previous Article in Special Issue
Investigation of the Presence of DNA in Human Blood Plasma Small Extracellular Vesicles
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Potential Pathogenic Impact of Cow’s Milk Consumption and Bovine Milk-Derived Exosomal MicroRNAs in Diffuse Large B-Cell Lymphoma

1
Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, D-49076 Osnabrück, Germany
2
University Clinic for Dermatology, Johannes Wesling Medical Centre, D-32429 Minden, Germany
3
Institute for Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, D-52074 Aachen, Germany
4
Institute of Nutrition, University of Giessen, D-35390 Giessen, Germany
5
Institute for Clinical Chemistry and Laboratory Medicine, University Hospital of Regensburg, University of Regensburg, D-93053 Regensburg, Germany
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(7), 6102; https://doi.org/10.3390/ijms24076102
Submission received: 26 January 2023 / Revised: 5 March 2023 / Accepted: 16 March 2023 / Published: 23 March 2023
(This article belongs to the Special Issue The Role of Exosomes in Health and Disease 2.0)

Abstract

:
Epidemiological evidence supports an association between cow’s milk consumption and the risk of diffuse large B-cell lymphoma (DLBCL), the most common non-Hodgkin lymphoma worldwide. This narrative review intends to elucidate the potential impact of milk-related agents, predominantly milk-derived exosomes (MDEs) and their microRNAs (miRs) in lymphomagenesis. Upregulation of PI3K-AKT-mTORC1 signaling is a common feature of DLBCL. Increased expression of B cell lymphoma 6 (BCL6) and suppression of B lymphocyte-induced maturation protein 1 (BLIMP1)/PR domain-containing protein 1 (PRDM1) are crucial pathological deviations in DLBCL. Translational evidence indicates that during the breastfeeding period, human MDE miRs support B cell proliferation via epigenetic upregulation of BCL6 (via miR-148a-3p-mediated suppression of DNA methyltransferase 1 (DNMT1) and miR-155-5p/miR-29b-5p-mediated suppression of activation-induced cytidine deaminase (AICDA) and suppression of BLIMP1 (via MDE let-7-5p/miR-125b-5p-targeting of PRDM1). After weaning with the physiological termination of MDE miR signaling, the infant’s BCL6 expression and B cell proliferation declines, whereas BLIMP1-mediated B cell maturation for adequate own antibody production rises. Because human and bovine MDE miRs share identical nucleotide sequences, the consumption of pasteurized cow’s milk in adults with the continued transfer of bioactive bovine MDE miRs may de-differentiate B cells back to the neonatal “proliferation-dominated” B cell phenotype maintaining an increased BLC6/BLIMP1 ratio. Persistent milk-induced epigenetic dysregulation of BCL6 and BLIMP1 expression may thus represent a novel driving mechanism in B cell lymphomagenesis. Bovine MDEs and their miR cargo have to be considered potential pathogens that should be removed from the human food chain.

1. Introduction

Diffuse large B-cell lymphoma (DLBCL) is a highly heterogeneous lymphoid neoplasm with variations in gene expression profiles, caused by genetic and epigenetic alterations. DLBCL is the most common type of non-Hodgkin lymphoma (NHL) worldwide, representing approximately 30–40% of all cases in different geographic regions [1,2]. Patients most often present with a rapidly growing tumor mass in single or multiple, nodal or extranodal sites [1,2]. Primary cutaneous diffuse large B cell lymphoma, leg type is an aggressive lymphoma with an inferior prognosis [3,4,5]. Recent DLBCL prevalence was estimated to be between 63,000 and 143,000 cases in the US [6]. The total number of incident and prevalent cases of DLBCL is expected to increase between 2020 and 2025 in the US and Western Europe [7]. The most popular classification by gene expression profiling subdivided DLBCL into three groups according to the cell-of-origin: the germinal center B-cell like (GCB) subtype, the activated B-cell like (ABC) subtype, and about 10–15% of cases being unclassifiable [2]. Patients with the GCB subtype usually have a better prognosis than patients with the ABC subtype [2,8,9]. Molecular pathology is complex and GCB and ABC tumors have different mutation and translocation profiles [2,10,11,12]. Gene mutations in DLBCL disturb various signaling pathways including histone modification, cell growth, proliferation, metabolism, differentiation, apoptosis, survival, homing/migration, response to DNA damage, B-cell receptor (BCR), signaling, Toll-like receptor (TLR) signaling, angiogenesis, and immunoregulation [10,13,14,15,16,17,18,19].
Among the well-established genetic aberrations, epigenetic deviations emerged as further important drivers of DLBCL lymphomagenesis [20,21,22,23,24,25,26,27,28]. Notably, the B cell lymphoma 6 (BCL6) proto-oncogene encodes a transcriptional repressor, which is required for germinal center (GC) formation and lymphomagenesis. Constitutive expression of BCL6 leads to DLBCL through activation-induced cytidine deaminase (AICDA)-mediated chromosomal translocations and mutations [10,29]. Recently, Jiao et al. [28] showed that AICDA and DNA methyltransferase 1 (DNMT1) induce BCL6 promoter methylation. Either loss of AICDA or DNMT1 with the instability of the AICDA-DNMT1 complex on the BCL6 promoter result in BCL6 promoter demethylation leading to increased BCL6 expression in DLBCL [28].
A variety of intrinsic and extrinsic pathogenic factors have been related to DLBCL pathogenesis. Prior radiation treatment, obesity, and smoking are most highly associated with DLBCL as well as infections with human immunodeficiency virus (HIV), Epstein–Barr virus (EBV), and human herpes virus 8 (HHV8) [30]. Drugs like phenytoin, digoxin, and TNF antagonists are also associated with lymphomagenesis as well as organic chemicals, pesticides, bisphenol A, phenoxy-herbicides, glyphosate, wood preservatives, dust, hair dyes, solvents, and prior chemotherapy [31,32,33,34,35,36,37,38].
Among dietary factors, total animal protein intake, meat, dairy, and milk consumption have been associated with an increased risk of NHL [39,40,41,42,43], whereas vegetable- and fruit-based diets reduce the risk of NHL including DLBCL [44,45,46,47,48]. Three recent large epidemiological studies discussed in the next chapter in more detail identified an increased risk of DLBCL by total dairy and cow’s milk consumption [49,50,51]. Cow’s milk is a major dietary exposure in industrialized countries often persisting over a lifetime [52]. Milk is not a simple nutrient, but operates as an endocrine signaling system of mammalian evolution enhancing mTORC1 activation [53,54].
It is the aim of this review to elucidate potential synergism in signal transduction between identified signaling pathways in DLBCL and milk-induced signaling pathways explaining the mechanistic link between cow’s milk consumption and DLBCL pathogenesis.

2. Epidemiological Evidence for Milk Intake and DLBCL Risk

In 2016, Wang et al. [49] performed a meta-analysis of 16 relevant articles related to dairy/milk consumption and NHL risk published up to October 2015. The pooled relative risks (RRs) (95% Confidence Intervals (CIs) of NHL for the highest vs. lowest category of the consumption of total dairy product, milk, butter, cheese, ice cream, and yogurt were 1.20 (1.02–1.42), 1.41 (1.08–1.84), 1.31 (1.04–1.65), 1.14 (0.96–1.34), 1.57 (1.11–2.20), and 0.78 (0.54–1.12), respectively. In subgroup analyses, the positive association between total dairy product consumption and the risk of NHL was found among case-control studies (RR = 1.41, 95% CI: 1.17–1.70, p = 0.368) but not among cohort studies (RR = 1.02, 95% CI: 0.88–1.17, p = 0.988). The pooled RRs of NHL were 1.21 (95% CI: 1.01–1.46, p = 0.140) for milk consumption in studies conducted in North America, and 1.24 (CI: 1.09–1.40, p = 0.245) for cheese consumption in studies that adopted validated food frequency questionnaires. A further NHL subtype analysis identified statistically significant associations between the consumption of total dairy product (RR = 1.73, 95% CI: 1.22–2.45, p = 0.67) and milk (RR = 1.49, 95% CI: 1.08–2.06, p = 0.33) and the risk of DLBCL. The dose-response analysis suggested that the risk of NHL increased by 5% and 6% for each 200 g/day increment in total dairy product and milk consumption, respectively [49].
In contrast, a systematic review and meta-analysis published in 2019 by Sergentanis et al. [55] found no association between milk and dairy product consumption and the risk of NHL based on three included studies published between 1996–2011 [44,56,57]. All of those reported on milk consumption. The pooled analysis pointed to a null association with NHL risk (pooled RR = 0.99, 95% CI: 0.85–1.15, p = 0.461) [55]. Conversely, the exposome-wide analysis within the European Prospective Investigation into Cancer and Nutrition study (475,426 participants) found 2,402 incident cases of B-cell lymphoma [51]. Notably, dairy product consumption was positively associated with B-cell lymphoma and DLBCL risk [51]. The prospective China Kadoorie Biobank study recruited ~0.5 million adults from ten diverse (five urban, five rural) areas across China during 2004–2008. A dairy intake of 50 g/day in urban regions exhibited an HR for lymphoma of 1.09 (95% CI: 0.89–1.34, p = 0.11), and for rural regions HR = 1.45 (95% CI: 1.07–1.96, p = 0.55) [50]. Taken together, recent epidemiological evidence points to an increased risk for NHL and in particular, DLBCL related to the consumption of milk and dairy products (Table 1).

3. Potential Milk-Related Factors Promoting DLBCL

3.1. Insulin-Like Growth Factor 1 Signaling in DLBCL

In the context of carcinogenesis, the most important functions of the insulin-like growth factor (IGF) family involve the intensification of proliferation and inhibition of cell apoptosis and effects on cell transformation through the synthesis of several regulatory proteins. The IGF axis controls survival and promotes metastases. Interactions of IGF axis components may be of a direct or indirect nature. The direct effects are linked to the activation of the PI3K/AKT/mTORC1 signaling pathway, stimulated by IGF-1 and IGF-1 receptor (IGF1R) activation. Activation of this signaling pathway increases mitogenesis and cell cycle progression but protects against different apoptotic stresses [58,59]. Sustained proliferation and evading apoptosis are critical hallmarks of cancers [60].
Recent evidence indicates that mitogenic IGF-1/IGF1R signaling plays a role in the pathogenesis of DLBCL [61,62,63,64,65]. DLBCL cell lines together with primary tumor cells derived from lymph nodes in four DLBCL patients were treated with the cyclolignan picropodophyllin (PPP), a selective inhibitor of IGF1Rs. PPP dose-dependently inhibited proliferation/survival in all DLBCL cell lines and primary cell preparations [61]. Remarkably, lower expression levels of Klotho, a physiological inhibitor of IGF1Rs [63], were observed in DLBCL patients and cell lines [64]. Enforced expression of Klotho could significantly induce cell apoptosis and inhibited tumor growth in DLBCL. Upregulation of Klotho resulted in declined activation of IGF1R signaling, accompanied by decreased phosphorylation of its downstream targets, including AKT and ERK1/2. Moreover, in a xenograft model treated with either Klotho overexpression vector or recombinant human Klotho administration presented restrained tumor growth and lower Ki67 staining [64]. Recently, Zhou et al. [65] observed aberrant activation of Hippo-YAP signaling in DLBCL. Loss of Hippo-Yes-associated protein (YAP) attenuated proliferation and induced cell cycle arrest in DLBCL cells [65]. Moreover, the downregulation of IGF1R expression led to a remarkable decrease in YAP expression. In contrast, exposure to IGF-1 promoted YAP expression and reversed the inhibition of YAP expression induced by IGF1R inhibitors [65]. IGF-1/IGF1R/YAP signaling may thus represent a new pathway in DLBCL tumorigenesis. Agarwal et al. [66] recently provided evidence that elevated levels of human oncoprotein Smoothened (SMO), a Frizzled-class-G-protein-coupled receptor, show a strong correlation with elevated levels of IGF1Rs and reduced survival in DLBCL patients. As an integral component of raft microdomains, SMO plays a fundamental role in maintaining high levels of IGF1Rs in lymphoma cells as well as IGF1R-associated activation of AKT (protein kinase B). Silencing of SMO increases lysosomal degradation and favors a localization of IGF1R to late endosomal compartments instead of early endosomal compartments from which much of the receptors would normally recycle. In addition, the loss of SMO interferes with the lipid raft localization and retention of the remaining IGF1Rs and AKT, thereby disrupting the primary signaling context for IGF1R/AKT. This activity of SMO is independent of its canonical signaling and represents a novel and clinically relevant contribution to signaling by the highly oncogenic IGF1R/AKT signaling axis [66]. Taken together, IGF-1/IGF1R signaling is involved in DLBCL tumorigenesis.

3.2. Milk-Induced IGF-1- and Amino Acid-Mediated mTORC1 Signaling

Regular consumption of commercial cow’s milk enhances circulating IGF-1 levels in children and adults [67,68,69,70,71,72,73,74,75,76]. There are two mechanisms leading to milk-mediated elevations of circulatory IGF-1 levels of the milk recipient: (1) Uncertain proportions of bovine milk IGF-1, which shares an identical amino acid sequence with human IGF-1 [77], may be absorbed in the human intestine. (2) Milk components, especially milk protein-derived amino acids (tryptophan, arginine, and methionine) may induce the synthesis and secretion of pituitary GH and hepatic IGF-1 into the circulation [78,79,80,81].

3.2.1. Milk-Derived Essential Branched-Chain Amino Acids

Milk proteins, especially whey proteins and caseins, are rich sources of essential branched-chain amino acids (BCAAs) that activate mTORC1 [54]. In all mammals except Neolithic humans, milk and milk protein exposure is restricted to the lactation period, whereas humans may be exposed to bovine milk proteins over their whole lifetime [52,82]. It is postulated that the type of protein in the diet influences directly the intrinsic capacity of the B lymphocytes to respond to an immunogenic stimulus [83]. The humoral immune response of mice fed with lactalbumin (20 g/100 g diet) was found to be higher than that of mice fed casein (20 g/100 g), soy protein (20 g/100 g) and wheat protein (20 g/100 g) diets, respectively [83]. To investigate the possible influence of dietary protein type on the supply of B lymphocytes, bone marrow lymphocyte production has been examined by a radioautographic assay of small lymphocyte renewal and an immunofluorescent stathmokinetic assay of pre-B cells and their proliferation [84]. The humoral response of all mice fed the lactalbumin-enriched diet was found to be higher than that of mice fed a casein diet or a control diet [84]. Cross and Gill [85] reported that bovine whey protein concentrate can modulate the proliferation of murine T and B cells in a dose-dependent manner. Among the major milk proteins, whey protein is the richest source of the essential BCAA leucine, which is the key amino acid activating mTORC1 via a RAG GTP-ase-dependent mechanism [86,87,88,89,90]. In contrast, reduced mTORC1 activity in B lymphocytes has been demonstrated at reduced extracellular amino acid and leucine concentrations [91]. The amount of leucine per g milk protein is a constant ratio for all mammals in the range of 10 g leucine/100 g milk protein [92]. Of all animal proteins, whey proteins contain the highest amount of leucine (14%) as compared to meat (8% leucine) [93]. Furthermore, in comparison to meat, whey proteins differ remarkably in their intestinal absorption kinetics, due to their fast intestinal hydrolysis increasing postprandial plasma amino acid levels [94,95,96,97].

3.2.2. L-Type Neutral Amino Acid Transporter 1 in DLBCL

L-type neutral amino acid transporter 1 (LAT1) is a heterodimeric membrane transport protein involved in the uptake of neutral amino acids such as leucine, isoleucine, valine, phenylalanine, tyrosine, tryptophan, methionine, and histidine [98]. Remarkably, recent evidence indicates that high LAT1 expression is a poor prognostic factor in NHL [99]. The LAT1 expression level in samples of NHL patients ranged from 1.9 to 99.2%, with a median of 42.4% (SD ± 29.5%). For the most prevalent subtypes, DLBCL had the highest median LAT1 expression (80.1%) [99]. The median LAT1 expression levels of the GCB type of DLBCL and non-GCB type were 70.4% ± 22.4 and 83.3% ± 11.6, respectively [99]. Of note, LAT1 expression correlated with Ki-67 expression and thus the proliferation of DLBCL cells [99]. Thus, persistent consumption of milk and milk proteins may boost LAT1/leucine/mTORC1-driven DLBCL proliferation.

3.2.3. Glutaminolysis in DLBCL

B-cell lymphomas use glutamine to power the tricarboxylic acid (TCA) cycle to generate energy and metabolic precursors [9,100]. Glutamine is metabolized through glutaminolysis to produce α-ketoglutarate. DLBCLs are dependent on mitochondrial lysine deacetylase sirtuin 3 (SIRT3) for proliferation, survival, self-renewal, and tumor growth in vivo regardless of disease subtype and genetics [101]. Importantly, overexpressed SIRT3 maintains DLBCLs’ metabolism by potentiating the TCA cycle through anaplerotic glutaminolysis [101]. In contrast, SIRT3 depletion impairs glutamine flux to the TCA cycle via glutamate dehydrogenase and reduces acetyl-CoA pools, which in turn induces autophagy and cell death [101]. Translational upregulation of activating transcription factor 4 (ATF4) is coupled with anaplerotic metabolism in DLBCLs due to nutrient deprivation caused by SIRT3 driving rapid flux of glutamine into the TCA cycle [102]. The active proliferation and high metabolic demand of DLBCL cells lead to a shortage of non-essential amino acids and results in translational activation of ATF4, which can transcribe target genes for the importation of extracellular nutrients to maintain the amino acid flux [102]. Moreover, the metabolic profile of DLBCL cells in the extracellular matrix is markedly different from cells in a suspension environment [103]. Recent evidence indicates that the synergistic consumption and assimilation of glutamine and pyruvate enables DLBCL proliferation in an extracellular environment-dependent manner [103].
Glutaminolysis is also important for the activation of mTORC1 [104,105]. mTORC1 is activated by glutamine and leucine via glutaminolysis-derived α-ketoglutarate upstream of RAG. This may provide an explanation for the glutamine addiction of cancer cells [105].

3.2.4. Milk Proteins: A Rich Source of Glutamine

Casein, the major protein fraction of cow’s milk and the major protein component of cheese, contains 9 g glutamine/100 g casein [106,107]. Thus, milk and dairy products, especially cheese, provide a rich source of glutamine, which may fuel DLBCL, enhancing TCA-driven tumor cell proliferation.

3.3. Activation of mTORC1 in DLBCL

mTORC1 serves as a rheostat that shapes differentiation along the B lineage, the pre-immune repertoire, and antigen-driven selection of mature B cells [91]. Aberrant and persistent activation of the PI3K/AKT/mTORC1 signaling pathway plays an important role in controlling the proliferation and survival of tumor cells in various types of malignancies, including DLBCL [108]. Aberrant and persistent activation of mTORC1 is often observed in malignant B cells such as NHL. Distinct mechanisms drive mTORC1 activation in the three most-common NHL types, i.e., DLBCL, follicular lymphoma, and mantle cell lymphoma. Constitutive activation of the B-cell receptor (BCR), PI3K, and TLR pathways is a hallmark of ABC-DLBCL [109]. Activation of these pathways is attributed to mutations in their components (most frequently CD79A/B, CARD11, TNFAIP3, and MYD88) and leads to chronic activation of NF-κB signaling, proliferation, and survival [110]. Within this context, the CBM (CARD11-BCL10-MALT1) complex acts as a supramolecular organizing center to set the activation threshold and amplify BCR signaling to the IKK complex and NF-κB [110,111,112]. Notably, it has recently been demonstrated in ABC-DLBCL cells that the MYD88, TLR9, and BCR form a supercomplex that co-localizes with mTORC1 on endolysosomes, where it drives pro-survival NF-κB and mTORC1 signaling [113]. Inhibitors of BCR and mTORC1 signaling cooperatively decreased the formation and function of the MYD88/TLR9/BCR supercomplex, providing mechanistic insight into their synergistic toxicity for MyD88/TLR9/BCR+ DLBCL cells [113]. In fact, mTORC1 activity is required to drive the increased expression of eIF4B, which is a feature of DLBCL [114]. Of note, the initiation factor of translation eIF4E, a downstream effector of mTORC1, has oncogenic effects in vivo and cooperates with C-MYC in B-cell lymphomagenesis [115]. Multi-level inhibition of the PI3K/AKT/mTORC1 signaling pathway in DLBCL showed significant anti-tumor effects in DLBCL [116,117,118,119,120,121,122,123,124]. Notably, rituximab combined with rapamycin synergically downregulated the PI3K/AKT/mTORC1 signaling pathway [118].

3.3.1. Milk-Induced Activation of mTORC1

Milk is an endocrine signaling system that promotes the activation of mTORC1 and mTORC1-dependent translation [53,54]. In comparison to a cow’s milk-free diet, young mice that had additional access to commercial cow’s milk exhibited increased expression of the mTORC1 downstream kinase pS6K1 in white adipose tissue and liver [125]. Remarkably, the mTORC1 response measured in mouse skeletal muscle following ingestion of high-quality plant-based and insect proteins was dampened compared to whey protein [126]. Bovine milk, a feeding and signaling system for postnatal anabolism and growth, activates IGF-1-driven activation of mTORC1 as well as BCAA- and glutamine-mediated activation of mTORC1. Persistent consumption of cow’s milk may thus augment mTORC1-mediated pathways augmenting mTORC1-driven DLBCL lymphomagenesis.

3.3.2. B-Cell Receptor Activation in DLBCL

The BCR signaling pathway is a crucial pathway of B cells, both for their survival and for antigen-mediated activation, proliferation, and differentiation [127]. Its activation is also critical for the genesis of many lymphoma types. BCR-mediated lymphoma proliferation may be caused by activating BCR-pathway mutations and/or by active or tonic stimulation of the BCR [128,129,130]. A substantial fraction of DLBCLs are addicted to oncogenic BCR and PI3K/mTORC1 signaling caused by different stimuli and various genetic aberrations [131,132]. It has been suggested that stimulation of BCR by specific antigens including autoantigens and antigens of infectious origin plays a pathogenic role in DLBCL [129,130]. Targeting BCR and downstream PI3K/AKT/mTORC1 signaling is a recent therapeutic approach in DLBCL [113,131,132].

3.4. Milk Peptide-Induced B-Cell Receptor Activation

Bovine milk proteins, whey proteins and caseins, are immunogens and allergens, even when proteins are present at very low concentrations. There are both conformational and linear epitopes, widely spread all along the protein molecules [133,134]. Bovine milk-derived peptides encrypting possible bioactive and/or immunogenic molecules originating from caseins, β-lactoglobulin, and minor milk proteins have recently been detected in human plasma after intake of pasteurized cow’s milk [135]. Cow’s milk protein allergy is a common condition encountered in young children [136,137], but may rarely also occur in adults [138]. Bovine milk proteins have been implicated to function as triggers for autoimmune diseases, especially type 1 diabetes mellitus [139,140,141,142,143], which has recently been linked to a moderately increased risk of NHL [144]. Cow’s milk protein sensitivity is related to irritable bowel syndrome in patients with primary Sjögren’s syndrome [145], which was associated with a 6.5-fold increased risk of NHL including DLBCL [146].
B-cell epitopes as a screening instrument for persistent cow’s milk allergy have been identified [147]. Immunoglobulin E (IgE) and IgG binding epitopes on β- and κ-casein have been detected in cow’s milk allergic patients [148]. Antibodies raised against peptide fragments of bovine α-s1-casein cross-react with the intact protein only when the peptides contain both B and T cell determinants [149]. Notably, bovine α-s1-casein and its peptides 61–110 and 91–110 contain both T and B cell determinants on α-s1-casein and can elicit peptide-native protein cross-reactive antibodies. Antibodies raised against peptide fragments of bovine α-s1-casein cross-react with the native protein but recognize sites distinct from the determinants on the protein [150]. Naïve mouse splenocytes stimulated with α- or κ-casein showed a similar immunogenic potential of both casein fractions. However, mice immunized with α-casein exhibited higher serum levels of IgG and IgA compared with mice immunized with κ-casein [151]. B-cell linear epitope analysis of milk proteins using in-silico tools showed κ-casein, β-casein, α-lactalbumin, β-lactoglobulin, αs1-casein, and αs2-casein contain 28, 36, 29, 28, 28, and 33 epitopes, respectively. Therefore, β-casein and αs2-casein have more B-cell epitope capacity [152].
Various physicochemical parameters can have an impact on the allergenicity of animal proteins [153]. Ultraheat treatment (UHT) of cow’s milk (100 °C/30 s) significantly altered the immunogenicity of most of the potent protein stimulants, which mostly coincided with their levels of protein denaturation. Pasteurization (72 °C/15 s) caused the least protein denaturation but altered the immunogenicity of several protein stimulants notably, including heat-stable caseins and α-lactalbumin [154]. In addition, it has been shown that heat treatment reduced the allergenicity of β -lactoglobulin by inducing conformational changes and by increasing its susceptibility to enzymatic digestion, both of which disrupted B-cell epitopes. Heat treatment alone did not alter the allergenicity of α-casein [155]. It is thus conceivable that raw and pasteurized milk in comparison to UHT and fermented milk exert stronger stimulatory effects on BCR signaling, which contributes to mTORC1 activation.

3.5. Estrogen Receptor-β Signaling in DLBCL

Recent evidence indicates that estrogens play a role in lymphomagenesis. According to the American Cancer Society Cancer Prevention Study-II Nutrition Cohort, a positive association between current postmenopausal combined use of estrogen and progestin and DLBCL has been observed [156]. Remarkably, increased expression of estrogen receptor-β (ERβ) has been detected in DLBCL cells [157,158,159]. Nuclear ERβ1 expression analysis in primary DLBCLs by immunohistochemistry revealed ERβ1 expression in 89% of the cases and was an independent prognostic factor for adverse progression-free survival in rituximab-chemotherapy treated DLBCL [157]. For nodal lymphoma, high ERβ expression (≥25%) was associated with poorer event-free survival independent of the international prognostic index with the adjusted hazard ratio (HR) of 2.49 (95% CI: 1.03–6.00, p = 0.042 [158]. ERβ is expressed at significantly higher levels in DLBCL compared to normal B cells, and ERβ plays a role in the protection against apoptosis in DLBCL [159]. Targeting of the ERβ with the selective estrogen receptor modulator tamoxifen reduced cell viability in all tested DLBCL cell lines [159]. In addition, tamoxifen-treated breast cancer patients showed a 38% reduced risk for DLBCL compared to breast cancer patients who did not receive tamoxifen [159].
As with estrogen receptor-α (ERα), estrogenic compounds including estrone (E1), 17β-estradiol (E2), and estriol (E3) activate ERβ. Relative to ERα, ERβ binds E3 and ring B unsaturated estrogens with higher affinity, while the reverse is true of E2 and E1 [160,161,162,163,164]. Interestingly, in lung cancer tissues and A549 cells, estrogen upregulated the IGF1R signaling through ERβ [165,166].

3.6. Milk-Derived Estrogens

Milk produced from “persistently” pregnant cows—the current routine praxis of the dairy industry to increase commercial milk yield—enhances milk estrogen concentrations. In pregnant cows, the predominant estrogen is E1 sulfate, which passes into milk [167]. Heat treatment (70 °C and 95 °C) does not affect E1 and E2 concentrations compared to unprocessed raw milk [168]. The concentration of E1 sulfate increases from 30 pg/mL in non-pregnant cows up to 151 pg/mL in pregnant cows at 40–60 days of gestation, and to a maximum level of 1000 pg/mL in cows at 220 days of gestation [169]. Farlow et al. [170] compared the estrogen concentrations of commercial cow’s and goat’s milk. They reported unconjugated and conjugated levels of E1 in regular whole cow’s milk of 129.9 ± 18.48 pg/mL and E2 levels of 28.19 ± 5.26 pg/mL, whereas in whole goat’s milk E1 and E2 levels were 42.48 ± 4.28 pg/mL and 17.87 ± 2.8 pg/mL, respectively [170]. The average milk concentrations of E1 (159 ng/kg) and E2 (6 ng/kg) in Swiss Holstein cows are comparable [171].
Pape-Zambito et al. analyzed E1 and E2 concentrations in pasteurized-homogenized milk and commercial dairy products [172]. E1 concentrations averaged 2.9, 4.2, 5.7, 7.9, 20.4, 54.1 pg/mL, and 118.9 pg/g in skimmed, 1%, 2%, and whole milks, half-and-half, cream, and butter samples, respectively. E2 concentrations averaged 0.4, 0.6, 0.9, 1.1, 1.9, 6.0 pg/mL, and 15.8 pg/g in skimmed, 1%, 2%, whole milks, half-and-half, cream, and butter samples, respectively [172]. The mean E2 mass in 237 mL of raw whole Holstein milk was 330 pg [173]. As the milk of pregnant dairy cows is pooled, commercial cow’s milk contains higher estrogen amounts compared to former times, when lactation of cows was synchronized and cows gave birth only in spring time. Maruyama et al. [169] analyzed the exposure to exogenous estrogen through the intake of commercial milk produced from pregnant cows in children and adults. Urine concentrations of E1, E2, E3, and pregnanediol significantly increased in all adults and children after intake of 600 mL/m2 of commercial cow’s milk. In prepubertal children, urinary excretion volumes of estrogens and pregnanediol significantly increased within 1–3 h. The net increase in E2 excretion from the basal E2 levels in urine (before the intake) was 39–109 ng/4 h in this study. These data indicate that the intake of estrogens from cow’s milk corresponds to the daily estrogen production rate in prepubertal boys [169]. Of note, not only milk but all dairy products, especially milk fat, contains estrogens that enhance the dietary estrogen exposure derived from cow’s milk and dairy products [174,175]. Milk products supply about 60–80% of ingested female sex steroids [174]. Taken together, it is conceivable that milk/dairy-derived estrogens via ERβ—IGF1R signaling may promote DLBCL cell proliferation.

3.6.1. Bisphenol A in Lymphomagenesis

Bisphenol A (BPA) is a common chemical used in the manufacture of materials in polycarbonate and epoxy plastic products and can interfere with the immune system. BPA is a proven endocrine disruptor capable of mimicking or blocking ERs and altering hormone concentrations and metabolism [176,177,178]. There is increasing concern that BPA perturbs the immune system causing adverse health effects including cancer [179,180,181]. It has recently been shown that BPA-induced DNA damages promote lymphoma progression in human lymphoblastoid cells through the aberrant catenin-β1 signaling pathway [37]. Gene-network analysis of microarray data sets in human lymphoma tissues as well as in human cells with BPA exposure to explore module genes identified potential pathways for lymphomagenesis in response to BPA [37]. BPA exposure resulted in a disrupted cell cycle and DNA damage by activating catenin-β1 encoded on the catenin-β1 gene (CTNNB1), the initiator of the aberrant constructed CTNNB1-NFKB1-AR-IGF-1-TWIST1 pathway, which may potentially lead to lymphomagenesis [37]. Activated catenin-β1 suppresses the DNA-repair-associated genes TP53 and CDKN1A [37]. Notably, dose-dependent colony formations of human lymphoblastoid TK6 cells visibly promoted by BPA treatment were significantly suppressed in siCTNNB1-transfected TK6 cells. In accordance, DLBCL showed a higher expression of β-catenin in contrast to reactive hyperplasia of lymph node tissues at both the mRNA and protein levels [182]. β-catenin is a key downstream effector of the Wnt/β-catenin pathway inducing the aging of mesenchymal stem cells through DNA damage response and the p53/p21 pathway [183]. Notably, constitutive activation of the DNA damage response pathway has been regarded as a novel therapeutic target in DLBCL [184]. In addition, increased TWIST expression, which is upregulated by IGF-1/IGF1R signaling [185], has also been observed in DLBCL [186]. Furthermore, canonical NF-κB signaling has been reported in DLBCL of the ABC type [187,188,189] and BPA is a known activator of NF-κB [190,191]. Of note, BPA also increases the expression of ERβ [192,193,194]. In thyroid tumor cells, BPA and E2 enhanced the expression of ERα/ERβ and GPR30 and activated AKT and mTORC1 [195].
Importantly, plasticizers like BPA modify the expression of oncogenic microRNAs (miRs) [196,197]. Notably, BPA exposure increased the expression of miR-21-5p in the majority of cell lines studied [196,197,198,199,200].

3.6.2. Contamination of Commercial Milk with Bisphenol A

BPA enters into the milk chain via multiple pathways at various points during milk production including PVC tubing used during the milking process, transfer from bulk milk to storage tanks, and during milk processing and packing [201,202]. Santonicola and colleagues reported mean BPA concentrations were 0.757 µg/L in manually milked samples, 0.580 µg/L in mechanically milked samples, and 0.797 µg/L in milk from the cooling tank [203]. Quantifiable levels were detected in samples obtained from the raw milk storage tank, pasteurized milk from the storage tank, and packaged milk. The highest BPA contamination levels were detected in raw milk from the storage tank (mean 0.265 µg/L) [204]. In milk samples (supplied in plastic bottles) of the winter season, BPA levels were 0.17–0.32 mg/kg, whereas, in milk samples of the summer season, BPA levels of 0.77–1.59 mg/kg have been detected [205]. BPA occurred in the milk chain as a result of different stages of milking and reached the highest levels at the end of the milk chain. Although the dietary intake of BPA is below the European Food Safety Authority’s temporary tolerable daily intake, exposure to BPA through milk consumption may have a critical impact on lymphomagenesis as BPA augments β1-catenin/ERβ/IGF-1/IGF1R-signaling involved in the pathogenesis of DLBCL (Figure 1).

3.7. Viral Agents in DLBCL

Viral agents are regarded as potential drivers of DLBCL tumorigenesis [206,207]. Various reports suggest an association of viral infections with DLBCL, including Epstein–Barr virus (EBV) also known as human herpes virus 4 (HHV4) [208,209,210,211,212,213,214,215,216], human immunodeficiency virus HIV (HIV) [217,218,219,220,221], hepatitis B virus (HBV) [222,223,224,225,226], hepatitis C virus (HCV) [227,228,229], human T-cell leukemia virus type 1 (HTLV-1) [230], and Simian virus 40 (SV40) infections [231,232,233], while human herpes virus 8 (HHV8) also known as Kaposi sarcoma-associated herpesvirus (KSHV) [30,234,235,236] and human papilloma virus (HPV) [237,238] associations with DLBCL are only rarely observed. EBV-positive DLBCL, not otherwise specified (NOS), is an EBV-positive clonal B cell lymphoid proliferation [239], that plays a major role in all virus-associated DLBCLs [214].
The oncogenic miR-155-5p is the most frequently upregulated miR in EBV-positive B cell malignancies. Of note, miR-155-5p plays a key role in B-cell immortalization by EBV [240]. EBV nuclear antigen 2 (EBNA2) and the B cell transcription factor interferon regulatory factor 4 (IRF4) are known to activate transcription of the host cell gene from which miR-155 is processed (miR-155HG; BIC). EBNA2 also activates IRF4 transcription, indicating that EBV may upregulate miR-155 through direct and indirect mechanisms [241]. Remarkably, miR-K12-11 encoded by KSHV shows significant homology to cellular miR-155, including the entire miR ‘seed’ region. Evidently, viral miR-K12-11 functions as an orthologue of miR-155 and probably evolved to exploit a pre-existing gene regulatory pathway in B cells. Moreover, the known etiological role of miR-155 in B cell transformation suggests that miR-K12-11 may contribute to the induction of KSHV-positive B cell tumors in infected patients [242].
High levels of B cell activation are induced by miR-21-5p in circulating B cells and are seen in HIV-infected individuals compared to non-infected controls. Notably, miR-21 is overexpressed in activated B cells of HIV-infected patients, suggesting its assistance in maintaining B cell hyperactivation contributing to lymphomagenesis [243,244]. Oncogenic viruses, including EBV, HBV, HCV, and HPV, co-evolve with their hosts and cause persistent infections. The upregulation of host miR-21 manipulates key cellular pathways to evade host immune responses and then promote viral replication [245]. Exosomes released by oncogenic virus-infected cells play a key role in promoting or inhibiting cancer formation [246].

3.8. Bovine Meat and Milk Factors

In recent years, a variety of circular replicase-encoding single-stranded (CRESS) DNA viruses and unclassified virus-like DNA elements have been discovered in a broad range of animal species and environmental samples. These sequences have been assigned to the virus phylum Cressdnaviricota, where CRESS stands for “Circular Rep-Encoding Single-Stranded”, to describe the functional features of these isolates, and viricota is the suffix for phylum taxa [247]. Especially, DNA elements termed bovine meat and milk factors (BMMF) are suspected to act as co-factors in the development of colon and breast cancer. BMMFs represent small single-stranded circular DNA, predominantly isolated from sera, milk, and dairy products of Eurasian cattle [248,249] and subsequently identified in periglandular cells of colon and breast cancers [250]. BMMF Rep protein has been detected in close vicinity of CD68+ macrophages in the interstitial lamina propria adjacent to colorectal cancer tissues, suggesting the presence of local chronic inflammation [251]. Recently, Nikitina et al. [252] found pleomorphic vesicles, regularly identified by staining peritumor tissues of colorectal, lung, and pancreatic cancer for expression of BMMF Rep. This subgroup of BMMF1 proteins is involved in the replication of small single-stranded circular plasmids of BMMF, but most likely also contributes to the formation of pleomorphic vesicular structures found in the periphery of colorectal, lung and pancreatic cancers [252]. These infectious agents share characteristics of both bacterial plasmids and known viruses [253]. Recently, circular single-stranded DNA genomes have been identified in the milk of sheep and goats [254] as well as the milk of water buffaloes [255]. Thus, dairy cows, sheep, goats, and water buffaloes add to the dispersal of CRESS viruses and circular ssDNA elements, which enter the human food chain via milk consumption [248,249,254,255]. According to de Villiers and Zur Hausen, future transcription analyses of acute lymphatic leukemias searching for BMMF-like infections will be of substantial interest [256]. These surveys should also include DLBCLs, the most common NHL linked to milk consumption (Table 2).

3.9. Bovine Leukemia Virus

Bovine leukemia virus (BLV) is the causative agent of leukemia/lymphoma in cattle. Cattle are the natural host of BLV, which integrates into B cells, producing a lifelong infection. BLV is a deltaretrovirus closely related to HTLV-1 and HTLV-2 [257] and causes enzootic bovine leukosis, which is the most important neoplastic disease in cattle [258]. Infectious BLV is present in the colostrum and milk of most BLV-positive cows [259,260]. Recent evidence points to a genetic flux between cattle and humans [261]. Olaya-Galán et al. [262] detected BLV DNA in raw beef and fresh milk for human consumption reminiscent of findings on BMMF distribution. Notably, BLV DNA was detected in the buffy coat cells of the blood of human subjects [263]. The most likely route of BLV transmission to humans is a foodborne infection [263]. After more than two decades, the opinion that “BLV does not infect humans” has started to change. BLV has been identified in human breast cancers giving rise to the hypothesis that it could be one of the causative agents of this condition [264,265]. Remarkably, one BLV miR, BLV-miR-B4, shares partial sequence identity and shares common targets with the host miR-29 [266]. Overexpression of miR-29 is associated with B cell neoplasms suggesting a possible contribution to BLV-induced tumorigenesis [266]. Bovine milk exosome-derived (MDE) miR-29 combined with BLV-derived miR-B4 may thus enhance oncogenic miR-29 signaling.

4. Exosomal MicroRNAs in the Pathogenesis of DLBCL

B cell development is a very orchestrated pathway that involves several molecules, such as transcription factors, cytokines, and miRs, respectively. All these components maintain the ideal microenvironment to control B cell differentiation. MiRs are small non-coding RNAs that target mRNAs to control gene expression. These molecules could circulate in the body in a free form, protein-bounded, or encapsulated into extracellular vesicles (EVs) including exosomes [267]. EVs represent a heterogeneous group of cell-derived membranous structures comprising exosomes and microvesicles, which originate from the endosomal system or which are shed from the plasma membrane, respectively. They are present in biological fluids and are involved in multiple physiological and pathological processes, especially cell-to-cell communication. Exosomes are small, nano-sized (50–100 nm) EVs secreted by cells and carry nucleic acids, proteins, lipids, and other bioactive substances that play a role in the body’s physiological and pathological processes. They are secreted by all cells and circulate in all body fluids including milk. Exosomes are key mediators of several processes in cancer that mediate tumor progression and metastasis. These nano-vesicles, when secreted from cancer cells, are enriched in non-coding RNAs (e.g., miRs) complexed with the RNA-induced silencing complex (RISC), that mediate an efficient and rapid silencing of mRNAs at the recipient cell, reprogramming their transcriptome [268]. The recently updated “hallmark of cancer model” outlined by Hanahan and Weinberg [269] considers the roles of miRs in non-mutational epigenetic reprogramming promoting cancer development and progression [269]. There is current interest in the role of miRs and their relevant targets in B cell development, B cell activation, and B cell malignant transformation [270]. B cell-specific miR expression plays a key role in the regulation of GC responses and B cell neoplasia [271,272]. The biological role of EVs, especially exosomes and their miR cargo in DLBCL initiation and progression is a recent focus of DLBCL research [273]. Rutherford et al. [274] showed that DLBCLs secrete large quantities of CD63-, ALIX-, TSG101-, and CD81-positive exosomes. Importantly, DLBCL cells take up exosomes and their RNAs [274]. This observation highlights the importance of EVs and exosomes promoting aberrant biological programming of recipient cells, including pre-metastatic niche formation and tumor progression [274,275], also demonstrated in other malignancies such as B-cell chronic lymphocytic leukemia [276], melanoma [277,278], pancreatic cancer [279], hepatocellular carcinoma [280,281], breast cancer [282,283], and prostate cancer [284,285]. Of note, CLL-derived microvesicles (MV) can activate the AKT/mTORC1/p70S6K/hypoxia-inducible factor-1α axis in CLL-bone marrow stromal cells (BMSCs) with the production of vascular endothelial growth factor, a survival factor for CLL B-cells. Moreover, MV-mediated AKT activation led to modulation of the β-catenin pathway and increased expression of cyclin D1 and C-MYC in BMSCs [275]. EVs and exosomes and their miR may play a key role in the pathogenesis of DLBCL, particularly with regard to the exchange of genomic information [273,274]. In fact, it has been observed that normal B cells internalize DLBCL-derived exosomes resulting in miR expression differences observed in normal B cells that are specific to lymphoma-subtypes [286].
Increased levels of exosomal miR-125b-5p and miR-99a-5p in DLBCL patients’ serum were associated with shorter progression-free survival time, and they can predict chemotherapeutic efficacy. Remarkably, DLBCL exosome-derived miR-125b-5p and miR-99a-5p were significantly upregulated and present predictive biomarkers for DLBCL chemotherapy resistance [287]. Exosomal miR-125b-5p is also described as a potential prognostic predictor of chemoresistance in the serum of patients with DLBCL [287]. Notably, exosomes carrying miR-125b-5p can reduce DLBCL sensitivity to rituximab by inhibiting the expression of tumor necrosis factor necrosis factor-α induced protein 3 (TNFAIP3), also known as A20 [288]. TNFAIP3 is a negative regulator of NF-κB that has been implicated as a tumor suppressor in multiple types of B-cell lymphoma including DLBCL [13,288,289,290,291,292]. A large percentage of DLBCL cases (55%) that have an MYD88 mutation also harbor a loss of TNFAIP3 [293].
B lymphocyte-induced maturation protein 1 (BLIMP1) also known as PR domain-containing protein 1 (PRDM1) plays a pivotal role in gene regulation required for B cell function and terminal differentiation and plasmablast formation [294,295]. BLIMP1 represses BCL6, a GC-restricted transcriptional repressor for GC formation [296,297,298,299]. BCL6 is a transcriptional repressor often expressed constitutively in DLBCL due to mutations of its genomic locus. BCL6 mediates aberrant survival, proliferation, genomic instability, and differentiation blockade in DLBCL cells [300,301,302]. In addition, BCL6 suppresses the expression of p53 and modulates DNA damage-induced apoptotic responses in GC B-cells. Of note, BCL6 represses p53 transcription by binding two specific DNA sites within the p53 promoter region and, accordingly, p53 expression is absent in GC B-cells where BCL6 is highly expressed [303]. Importantly, BCL6 overexpression represses BLIMP1, suppressing plasma cell differentiation [304].
The majority of TP53 mutations in human DLBCL are accompanied by loss of p53 function [305]. Pascual et al. [306] recently demonstrated that DNA damage response by p53 is a central mechanism suppressing the pathogenic cooperation of IKK2ca-enforced canonical NF-κB and impaired differentiation resulting from BLIMP1 loss in ABC-DLBCL lymphomagenesis. Conditional deletion of p53 in mouse GCBs strongly synergized with IKK2 activation and BLIMP1 loss to promote GC-derived lymphomagenesis. Thus, there is a close interaction between BLIMP1, BCL6, and p53 in the pathogenesis of DLBCL. BLIMP1 has been substantiated as a key tumor suppressor of DLBCL [307,308,309,310,311,312,313,314,315]. Mandelbaum et al. [313] and Calado et al. [314] unequivocally demonstrated that BLIMP1 functions as a tumor suppressor and guardian of ABC-like DLBCL lymphomagenesis. Loss of BLIMP1 function contributes to the overall poor prognosis of ABC-DLBCL patients [316,317]. Furthermore, dysregulated multidrug resistance protein 1 (MDR1; ABCB1) by BLIMP1 is involved in the doxorubicin resistance of non-GC B-cell-like DLBCL [318].
Notably, loss of BLIMP1 was associated with MYC overexpression and decreased expression of p53 pathway molecules [317]. BLIMP1 represses C-MYC promoter activity in a binding site-dependent manner [319]. The repression of C-MYC by BLIMP1 is a critical step for terminal B cell differentiation [319]. The upregulation of C-MYC gene expression induced by PRDM1 inactivation apparently plays a crucial role in the development of DLBCL [320].

4.1. MicroRNA-Mediated Transcriptional Regulation in DLBCL

BLIMP1 has been identified as a key immune gene hub regulated by miRs [321]. Inactivation of BLIMP1 not only results from PRDM1 mutations but also from miR-mediated downregulation of BLIMP1 expression. Ma et al. [322] demonstrated that BLIMP1 suppression via EBV-miR-BHRF1-2 plays a potential role in EBV-induced lymphomagenesis. Recently evidence indicates that certain miRs act as oncogenes in DLBCL pathology [323]. In the following section pivotal DLBCL-related miRs that are also signature miRs of cow’s milk and milk exosomes will be discussed in more detail.

4.1.1. MicroRNA-let-7 Over-Expression in DLBCL

High levels of let-7a and miR-9 in HL cell lines correlated with low levels of BLIMP1 [324]. Let-7, in particular let-7b, is overexpressed in DLBCL relative to normal GC B-cells, suggesting that this miR is deregulated. Thus, abnormal epigenetic downregulation of BLIMP1 by let-7 and other miRs may represent an alternative epigenetic mechanism reducing BLIMP1 function in a subset of DLBCL [325].

4.1.2. MicroRNA-125b Over-Expression in DLBCL

Malumbres et al. [326] provided experimental evidence that GC-enriched hsa-miR-125b down-regulates the expression of IRF4 and BLIMP1. Dysfunction of TP53, including miR regulations, copy number alterations of the p53 pathway and p53 itself, dysregulation of p53 regulators, and somatic mutations by abnormal p53 function modes, play an important role in DLBCL generation, progression, and invasion [327]. Importantly, miR-125b-5p is a key negative regulator of p53 expression and targets the 3′ untranslated region (3′ UTR) of TP53 mRNA [328,329]. Li et al. [330] reported mutations in the 3′ UTR of TP53 mRNA that modify miR-125b binding and the expression of p53 affected the response to therapy in DLBCL patients. Furthermore, miR-125b-5p targets MAX dimerization protein 4 (MXD4) thereby enhancing C-MYC activity [331].

4.1.3. MicroRNA-21 Over-Expression in DLBCL

Elevated levels of miR-21-5p have been found in the serum of patients with DLBCL [332,333,334,335,336,337,338]. miR-21-5p contributes to characteristic miR signatures in DLBCL [287,339,340,341]. High miR-21-5p expression is associated with the ABC subtype of DLBCL [340]. miR-21-5p is the most frequently deregulated miR in malignancy, including B-cell lymphomas, and it has oncogenic potential downstream of STAT3 [342]. Remarkably, BLIMP1 binds to the promoter of pri-miR-21 and represses pri-miR-21 expression [342]. Using in situ hybridization, miR-21 expression was also detected in the stromal compartment of 73.2% of DLBCL [343]. DLBCL patients with higher miR-21 expression have shown significantly worse overall survival than those with lower miR-21 expression [335]. Inada et al. [334] confirmed that miR-21-5p is not only increased in serum but also in exosome-enriched serum of patients with DLBCL. Circulating exosomal miRs can be useful noninvasive biomarkers for the diagnosis of DLBCL and for improving the identification of patients with poor outcomes [344].
Whereas C-MYC induces the expression of miR-21, miR-21 in turn targets MAX dimerization protein 1 (MXD1) mRNA and downregulates MXD1 protein. The resultant decrease in MXD1 promotes the formation of a C-MYC-MAX heterodimer, leading to sustained C-MYC activation [345]. Thus, C-MYC/miR-21/MXD1 represents a positive-feedback loop that is critical for the maintenance of B-cell lymphoma survival. Targeting miR-21 with the bendamustine derivative NL101 blocked the C-MYC/miR-21/MXD1 loop, a potential therapeutic strategy of C-MYC-directed lymphoma therapy [345]. Von Hippel-Lindau (VHL) mRNA is another direct target of miR-21-5p in DLBCL [346]. In contrast, curcumin is a known suppressor of miR-21 [347] and decreases miR-21 levels through both increasing miR-21 exosome exclusion from the cells and inhibiting the transcription of the miR-21 gene (MIR21) in the cells by binding to its promoter [347]. In fact, curcumin exerted its anti-proliferation, anti-migration, anti-invasion, and pro-apoptosis functions, at least partly, by repressing miR-21 and regulating VHL expression in the DLBCL cell line [346]. Furthermore, miR-21-5p inhibits phosphatase and tensin homolog (PTEN) and programmed cell death protein 4 (PDCD4), thus increasing PI3K/AKT-mediated cell proliferation and cell survival as well as eIF4A and eIF4G-mediated translation [347]. In addition, miR-21 via activation of AKT increases NF-κB signaling [347,348,349]. miR-21 level was inversely correlated with the levels of FOXO1 and PTEN in DLBCL cell lines [350,351]. Reporter-gene assay showed that miR-21 directly targeted and suppressed FOXO1 expression, and subsequently inhibited BCL2-like 11 (BIM) transcription in DLBCL cells. miR-21 also down-regulates PTEN expression and consequently activates the PI3K/AKT/mTORC1 pathway, which further decreases FOXO1 expression. In contrast, FOXO1 activation is a required effector for spleen tyrosine kinase (SYK) and AKT inhibition in tonic BCR signal-dependent DLBCL [352]. Moreover, miR-21 inhibitor suppressed the expression and activity of multidrug resistance protein 1 (MDR1), thereby sensitizing DLBCL cells to doxorubicin [350]. In fact, miR-21 exhibited significantly higher plasma levels in patients with DLBCL unresponsiveness to treatment [353], whereas inhibition of miR-21 induced suppression of proliferation and invasion, as well as increased apoptosis in DLBCL [354]. According to a recent meta-analysis, poor overall survival has been associated with high expression of miR-21 in tumor tissue of DLBCL [355]. Recently, a novel prognostic model based on four circulating miRs (miR-21, miR-130b, miR-155, and miR-28) in DLBCL with implications for their roles of myeloid-derived suppressor cells (MDSC) and Th17 cells in lymphoma progression [356]. Remarkably, inhibition of miR-21 reduced the expression of IGF-1 [356]. The high-risk group of the 4-circulating miRs prognostic model was characterized by activation of RAS protein signal transduction. Both IGF-1 and JUN were two key regulators of the RAS cascade, enhancing lymphoma development [356]. As a mechanism of action, IGF-1 and JUN are positively regulated by miR-21 [356]. There is recent interested in miR-based therapies in B-cell NHL [357].

4.1.4. Over-Expressed MicroRNA-155 in DLBCL

The evolutionarily conserved miR-155 plays an important role in the immune system, specifically in regulating GC reaction to produce an optimal T cell-dependent antibody response [358]. miR-155-5p and miR-155-3p are processed from the B-cell integration cluster (BIC) gene (now designated, MIR155 host gene or MIR155HG). miR-155-5p is highly expressed in both activated B and T cells and monocytes/macrophages and proliferating lymphoblastoid cell lines [240,359]. miR-155 can be processed from sequences present in BIC RNA, a spliced and polyadenylated but non-protein-coding RNA that accumulates in lymphoma cells [360]. Isolates of several types of B-cell lymphomas, including DLBCL have 10- to 30-fold higher copy numbers of miR-155 than do normal circulating B cells [360]. Significantly higher levels of miR-155 are present in DLBCLs with an ABC phenotype than with the GC phenotype [360]. In fact, oncogenic miR-155 is highly expressed in the non-GC B-cell or activated B-cell subtype of ABC-DLBCL [361]. In accordance, miR-155 expression levels in formalin-fixed/paraffin-embedded tissue of patients with DLBCL were significantly higher in de novo DLBCL patients compared to controls [362]. PI3K is activated in DLBCL cells due to chronic or tonic BCR signaling [363]. miR-155 is potentially involved in the upregulation of BCR-PI3K-AKT-mTORC1 signaling DLBCL. Huang et al. [364] reported a link between miR-155 overexpression in DLBCL and overactivated PI3K-AKT signaling. Notably, the PI3K regulatory subunit 1 (PIK3R1; also known as p85α), which is a negative regulator of the PI3K-AKT pathway, is a direct target of miR-155-5p and miR-21-5p [364]. Knockdown of miR-155 in OCI-Ly3 cells diminished AKT activity [364]. PTEN has been identified as a further target of miR-155 [365,366]. It has recently been reported that miR-155 represses the mTOR phosphatase DEP domain-containing protein 6 (DEPD6) also known as DEP domain-containing mTOR-interacting protein (DEPTOR) as well as C-CBL (SYK ubiquitin E3 ligase) [367]. DEPTOR protein expression was markedly lower in more aggressive non-GCB DLBCLs than in GCB tumors [367]. DEPTOR is capable of inhibiting the kinase activity of mTOR of both mTORC1 and mTORC2 [368,369]. AKT Thr308 being phosphorylated by phosphoinositide-dependent kinase 1 (PDK1), the plasma membrane-localized and activated mTORC2 further phosphorylates AKT at Ser473 [370]. Thus, miR-155 upregulates PI3K-AKT-mTORC1 signaling at multiple inhibitory checkpoints. Upregulated miR-155, mTORC1, pS6K1, and downregulated DEPTOR were associated with the treatment-resistant group in gastric DLBCL [371]. The inositol polyphosphate-5-phosphatase (INPP5D also known as SHIP1) is another target of miR-155-5p [372]. SHIP dephosphorylates the PI3K product and lipid second messenger phosphatidylinositol-3,4,5-trisphosphate [PI(3,4,5)P3] to produce phosphatidylinositol-3,4-bisphosphate [PI(3,4)P2] [373,374]. SHIP plays a critical role in the termination of PI [3,4,5]P(3) signals that follow BCR aggregation [374]. B-cell precursors from SHIP-deficient mice progress more rapidly through the immature and transitional developmental stages and SHIP-deficient B cells have increased resistance to BCR-mediated cell death [374]. Diminished SHIP1 expression in DLBCL resulted in autocrine stimulation by tumor necrosis factor-α (TNFα) [375]. Whereas miR-155 downregulates SHIP expression increasing PI3K-AKT signaling, a novel SHIP1 activator, AQX-435, reduced AKT phosphorylation and growth of DLBCL in vivo and cooperated with ibrutinib for tumor growth inhibition [376]. DLBCL cases with elevated circulating levels of miR-155 had shorter overall survival than those with a lower miR-155 expression [377]. Notably, a significant increase in plasma exosomal miR-155, let-7g, and let-7i levels and exosome concentration in refractory/relapsed patients compared to responsive patients and patients receiving R-CHOP has been reported [378]. In contrast, the inhibition of miR-155 by cobomarsen, an oligonucleotide inhibitor of miR-155, slowed DLBCL tumor cell growth [360]. Selective inhibition of miR-155 function specifically inhibits the growth of lymphoblastoid cell lines and the DLBCL cell line IBL-1 [240]. Chronic active BCR signaling plays a pathogenetic role in ABC DLBCL [14]. The activation of ‘chronic’ or ‘tonic’ BCR signaling in lymphoma B cells can be influenced by a specific immunoglobulin structure, the expression and mutations of adaptor molecules (like GAB1, BLNK, GRB2, and CARD11), the activity of kinases (like LYN, SYK, and PI3K) or phosphatases (like SHIP-1, SHP-1, and PTEN) and levels of oncogenic miRs, predominantly miR-155-5p [379].

4.1.5. MicroRNA-148a Maintains Survival of Immature B Cells

miR-148a has been identified as a critical regulator of B cell tolerance and autoimmunity [380]. Elevated miR-148a-3p expression impaired B cell tolerance by promoting the survival of immature B cells after engagement of the BCR by suppressing the expression of growth arrest- and DNA-damage-inducible gene α (GADD45A), the PI3K inhibitor PTEN, the pro-apoptotic protein BIM as well as the TNF receptor subfamily, member 1B (TNFRSF1B) [380]. Of note, TNFRSF1B is also a target of miR-125b-5p and let-7-5p. GADD45α regulates G2/M checkpoints and the entry to mitosis by dissociating the CDK1/cyclin B kinase complex [381]. Moreover, GADD45α participates in DNA repair by binding and activating the proliferating cell nuclear antigen, a key element in excision base repair [382]. Interestingly, apoptosis induced by anti-MEK small molecule AZD6244 was BIM-dependent in DLBCL cells [383].
Recent evidence indicates that miR-148a-3p promotes plasma cell (PC) differentiation via targeting GC transcription factors BTB and CNC homology 2 (BACH2) and microphthalmia-associated transcription factor (MITF) [384]. For differentiation of mature activated B cells into long-lived antibody-secreting PCs, BACH2 and MITF, are essential, as they delay premature differentiation of GC B-cells by repressing BLIMP1 and IRF4 [384]. Therefore, BACH2 and MITF expression must be attenuated in activated B cells to allow terminal PC differentiation. Notably, high BACH2 expression in GCB and non-GCB DLBCL exhibited shorter 3-year overall survival compared to DLBCLs with low BACH2 expression [385]. The analysis of miR-148a-deficient mice revealed reduced serum Ig, decreased numbers of newly formed plasmablasts, and reduced CD19-negative, CD93-positive long-lived plasma cells [386].
Thus, miR-148a may exert dual functions in B cell development, i.e., promoting the survival of immature B cells but also driving terminal differentiation of PCs. According to a recent systematic review by Yazdanparast et al. [273], exosomal miRs are not only involved in critical pathobiochemical mechanisms in DLBCL but are also useful for diagnosis and prognosis in DLBCL and are promising therapeutic tools and predictors of response to therapy.

4.2. Potential Uptake of Bovine Milk-Derived Exosomes by B Cells

Pasteurized milk is consumed in high quantities in the US and northern European countries. Swedes, for instance, who per capita consumed 98 L milk in 2018, primarily drink pasteurized milk [387]. In contrast to ultraheat treatment (UHT) (135–150 °C, 1–10 s), pasteurization (72 °C for 15 s) is a gentle thermal processing [388]. Pasteurization in contrast to UHT preserves milk exosomes and substantial amounts of their miRs [389,390]. Compared to raw cow’s milk, pasteurization and homogenization reduces the concentration of milk EVs [389]. However, it should be taken into account that cow’s milk contains 1012–1014 exosomes per mL [391] and thus delivers 10- to 1000-fold higher exosome numbers/mL compared to human milk. Bovine milk exosomes survive the harsh conditions in the gastrointestinal tract [392,393].
Bovine milk-derived exosomes (MDEs) are taken up by endocytosis as shown in intestinal and vascular epithelial cells [394,395] and breast cancer cells [396]. There is compelling evidence that bovine MDEs and their miRs are bioavailable [397,398,399,400], reach the systemic circulation [387], and enter the cells and peripheral tissues [401,402,403,404,405,406,407,408]. There is ample evidence that human and bovine MDEs are taken up by human peripheral blood mononuclear cells (PBMCs) [409], including macrophages [410,411,412] and T cells [413]. Wolf et al. [394] showed that the uptake of bovine MDEs is mediated by endocytosis and depends on cell and exosome surface glycoproteins in human and rat intestinal cells. In accordance, Kusuma et al. [395] observed that bovine MDEs are taken by endocytosis in vascular endothelial cells. Curcumin encapsulated in MDEs resists human digestion and possesses enhanced intestinal permeability in vitro [414]. Recently, González-Sarrías et al. [396] reported the uptake of curcumin and resveratrol-loaded bovine MDEs in MCF-7 breast cancer cells primarily was dependent on clathrin-mediated endocytosis. Caner et al. [286] recently showed that normal B cells are able to internalize exosomes derived from DLBCL resulting in changes in miR expression profiles specific to lymphoma subtypes. B cells exhibit a very active endocytic machinery. The best-characterized mechanism of BCR internalization is clathrin-mediated endocytosis [415,416,417,418,419,420]. Recent evidence indicates that class A scavenger receptor-1/2 facilitates the uptake of bovine MDEs in murine bone marrow-derived macrophages and C57BL/6J mice [421]. Baos et al. [422] described the presence of class A scavenger receptor 1 (CD204) in several cell populations of PBMCs, specifically in T and B lymphocytes, as well as monocytes. Notably, an increase in CD204+ cell numbers was associated with poor clinical outcome in DLBCL of the central nervous system [423]. Betker et al. [402] suggest that the absorption of bovine MDEs from the gastrointestinal tract is mediated via neonatal Fc receptor (FcRn). Of note, the expression of FcRn has been demonstrated on human B cells and murine primary B cells [424,425,426]. Thus, several potential mechanisms may mediate the uptake of bovine MDEs by B cells and B cell precursors.

4.3. Bovine Milk Exosome-Derived MicroRNAs

4.3.1. MicroRNAs of the Let-7 Family

The most abundant miRs of MDEs are highly conserved between mammals [427,428]. Interestingly, several abundant miRs including the let-7 family members let-7a, let-7b, let-7f, and miR-148a are shared between species. Moreover, milk-derived miRs have been implicated in immune-related functions and regulation of cell growth and signal transduction [428,429]. Interestingly, the bta-let-7 family members let-7a, let-7b, let-c, let-7d, let-7f, let-7g, and let-7i are expressed in bovine MDEs [411,428,429,430,431,432,433,434]. Notably, in vivo treatment with bovine MDEs resulted in an increase in let-7a and miR-148a and expression in hepatic stellate cells [435]. Thus, the potential uptake of bovine MDEs by DLBCL cells may enhance cellular let-7 abundance promoting epigenetic down-regulation of BLIMP1 [325].

4.3.2. MicroRNA-125a and MicroRNA-125b

A large quantity of bovine milk bta-miR-125b (∼109–1010 copies/300 mL milk) withstood digestion under simulated gastrointestinal tract conditions [383]. Of note, bovine milk bta-miR-125b that resists digestion was associated with exosomes [411]. However, the majority of digested dairy milk bta-miR-125b was associated with EVs sedimenting at a centrifugation speed lower than that for exosomes [392]. Comparative expression profiles of immune-related miRs of buffalo milk nanovesicles (50–200 nm) confirmed the expression of miR-125b, miR-155, and miR-21, respectively [436]. Notably, both human and bovine miR-125b-5p exhibit an identical sequence (mirbase.org). Importantly, miR-125b targets TP53 [329,330] and TNFAIP3 (A20) [288]. TNFAIP3 (A20) is a tumor suppressor gene in lymphomas [188,437] and is frequently inactivated in B-cell lymphomas [438]. A20 inhibits NF-κB signaling [439,440,441]. Evidence has been presented that epigenetic termination of TNFAIP3 function by miR-125a and miR-125b could strengthen and prolong NF-κB activity, which promotes DLBCL lymphomagenesis [442]. Shome et al. [443] identified bta-miR-125a as one of the most abundant miRs of commercial milk related to immunological roles that were not affected by pasteurization, homogenization, or heat treatment. The seed sequence of hsa-miR-125a-5p and bta-miR-125a-5p are identical. The fine-tuning of TNFAIP3 levels mediated by miR-125 expression had a striking impact on K-63 ubiquitination of TRAF2 and RIP1, IκBα degradation, p65 nuclear accumulation, and transcription of NF-κB target genes [443]. These events defined an antiapoptotic profile compatible with constitutive NF-κB activation and enhanced the fitness of B lymphoma cells.
Together, miRs of the miR-125 family play a key role in immune cell activation and oncogenesis, and constitutive activation of the NF-κB pathway in DLBCL [442,443,444]. In accordance, exosome-derived miR-125b-5p reduced the sensitivity of DLBCL to chemotherapy and rituximab [277,278]. Furthermore, the miR-125 family has been implicated as an aggravating factor in several other B cell malignancies [444].

4.3.3. MicroRNA-21

Bovine exosomal miR-21 was not affected by digestion in vitro [396] and was stable under different household storage conditions, indicating that miR-21 could be biologically available to milk consumers [436]. Notably, human and bovine miR-21-5p share identical nucleotide sequences [445]. Mutai et al. [446] confirmed miR-21-5p as a component of exosomes derived from pasteurized commercial cow’s milk and showed that miR-21-5p plasma levels significantly increased by 147% 3.2 h after consumption of 1 L commercial milk by healthy human volunteers [446]. miR-21 is an abundant signature miR of cow’s milk [427,430,447] and is a component of bovine MDEs [268,446,448]. In addition to miR-125b-5p, miR-21-5p targets and inhibits TNFAIP3 [449,450]. Furthermore, miR-21 plays an oncogenic role by targeting FOXO1 [451,452,453] and PTEN [454,455,456], thereby activating the PI3K/AKT/mTORC1 pathway in DLBCL [350]. B-cell lymphoma/leukemia 11B (BCL11B) is another direct target of miR-21 [457]. Its loss of function in mice contributes to lymphomagenesis [458,459]. Activation of eIF4F by increased mTORC1 signaling has a direct role in lymphomagenesis due to increased synthesis of oncogenes that are dependent on enhanced eIF4A RNA helicase activity for translation [460]. AKT activation leads to the degradation of programmed cell death 4 (PDCD4), which inhibits the translation initiation factor EIF4A, an RNA helicase that catalyzes the unwinding of secondary structure at the 5′ UTR of mRNAs [461] and can alter eIF4F complex formation [460]. Notably, PDCD4 is also epigenetically downregulated by miR-21 [239,354,355]. It is well-accepted that miR-21 is frequently upregulated in DLBCL [342,343,344]. Moreover, insulin-like growth factor (IGF)-binding protein-3 (IGFBP3) is a miR-21 target gene promoting glioblastoma tumorigenesis [462]. Thus, miR-21 enhances levels of free IGF-1. The majority of reported targets of miR-21 are tumor suppressor genes and inhibitors of apoptosis [239,463,464,465] (Figure 2).

4.3.4. Exosomal MicroRNA-21 Exposure and M2 Macrophage Polarization

The tumor microenvironment (TME) of DLBCL and its role in DLBCL pathogenesis and patient survival is a matter of recent research [466]. The TME consists of T and B lymphocytes, tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), cancer-associated fibroblasts (CAFs), and other components [467]. The crosstalk between malignant B cells and immune cells in the lymphoma TME is highly complicated and might be affected by often interconnected intrinsic and/or extrinsic mechanisms that ultimately can lead to immune escape [467]. M2-polarized macrophages are implicated to contribute to DLBCL progression and poor patient outcome [466,468]. Tumor-associated macrophages (TAMs) comprise an important part of the TME, are predominantly M2-polarized, and play a key role in DLBCL progression [469,470,471,472]. Compelling evidence in various cancer entities underlines the impact of miR-21-5p in driving M2 polarization of macrophages and TAMs [473,474,475,476], whereas inhibition or depletion of miR-21 reversed M2 polarization back to the M1 phenotype [477,478,479]. Remarkably, miR-21 depletion in macrophages promotes tumoricidal polarization and enhances PD-1 immunotherapy [477].
Exosome-mediated crosstalk between tumors and TAMs involves the traffic of miR-21, which is the recent focus on molecular oncology [480]. DLBCL-derived EVs and exosomes are internalized by macrophages and can induce M2 macrophage polarization and thus contribute to tumor progression [481,482]. Importantly, tumor-cell-derived EVs and exosomes transfer miR-21-5p to monocyte/macrophages inducing the M2-phenotype [483,484,485,486,487]. In fact, it has been demonstrated that miR-21-5p-containing exosomes were engulfed by CD14+ human monocytes, suppressing the expression of M1 markers and increasing that of M2 markers [484]. The transcription factor STAT3 is a known enhancer of miR-21 expression [488,489,490]. Preclinical evidence indicates that the STAT3 inhibitor pacritinib could overcome temozolomide resistance via downregulating miR-21-enriched exosomes from M2 glioblastoma-associated macrophages [491]. It has been demonstrated in colon cancer that TAM secrete exosomes containing miR-21-5p and miR-155-5p promoting cell migration and invasion [492]. In hepatocellular carcinoma (HCC), miR-21-5p expression was upregulated in M2 macrophage-derived EVs, which carried miR-21-5p into HCC tissues [493]. Of note, M2 macrophage-derived EVs promoted the depletion of CD8+ T cells in HCC via the miR-21-5p/YOD1/YAP/β-catenin axis [493]. In pancreatic cancer, M2 macrophage-derived exosomal miR-21-5p stimulated differentiation and activity of pancreatic cancer stem cells via targeting Kruppel-like factor 3 (KLF3) [494].
Taken together, compelling evidence supports the view that tumor cell-derived exosomal miR-21-5p promotes macrophage M2 polarization within the TME. M2-polarized TAMs themselves secrete miR-21-5p enriched exosomes into the TEM further stimulating tumor cell growth. In this scenario, it is of critical concern that bovine MDEs, which are enriched in miR-21-5p [227,446,448] are taken up by human PBMCs [403] and human macrophages [411]. Thus, dietary MDE miR-21 exposure may enhance the total burden of miR-21 signaling in the TME promoting lymphomagenesis and tumor progression [334] (Figure 3).

4.3.5. MicroRNA-29b

In comparison to colostrum, mature cow’s milk contains more abundant amounts of miR-29b [411]. Pasteurized and homogenized 2% fat commercial cow’s milk stored at 4 for 15 days still exhibited more than 50% of the initial miR-29b concentration detected in fresh raw cow’s milk [495]. Yu et al. [448] reported that bovine MDEs contain miR-21 and miR-29b, whereas Izumi et al. [411] detected miR-29c in bovine MDEs. Baier et al. [409] demonstrated that postprandial plasma concentrations of miR-29b increased in a dose-dependent manner after intake of 0.25, 0.5, and 1.0 L of commercial cow’s milk in healthy human volunteers. Notably, the expression of runt-related transcription factor 2 (RUNX2), which is positively regulated by miR-29b [496], increased by 31% in PBMCs after milk consumption compared with baseline [409]. Of note, the nucleotide sequence of bovine miR-29b is identical to that of human miR-29b [497]. RUNX2 is highly expressed in adherent B-NHL cells compared to cells in suspension, and knockdown of RUNX2 expression could reverse cell adhesion-mediated drug resistance. Furthermore, RUNX2 could promote the proliferation of B-NHL cells [498]. Hines et al. [499] reported that miR-29s in murine B lymphocytes regulate the BCR-PI3K signaling cascade by dampening PTEN expression and that loss of this miR cluster results in increased apoptosis as well as defects in B cell terminal differentiation. Remarkably, the miR expression profile of U2932 DLBCL cells transfected with EBV-encoded latent membrane protein 1 (LMP1) exhibited significant upregulation of miR-29a, miR-29b, and miR-29c [500]. However, in this experimental model system of DLBCL, miR-29b downregulated the expression of the T-cell leukemia gene 1 (TCL1) [501], which exhibits oncogenic activity in distinct classes of B cell lymphoma [501,502].

4.3.6. MicroRNA-155

The quantity of immune-related miR-155 of colostrum is 5.4-fold higher than in raw mature cow’s milk [429]. Levels of miR-155 of HTST (high temperature for short time, 75 °C for 15 s) milk was 3.8-fold higher for miR-155 compared to LTLT (low temperature for a long time, 63 °C for 30 min) cow’s milk [429]. The lowest miR-155 levels have been detected in UHT (ultra-heat treatment 120–130 °C for 0.5–4 s) [429]. It appears that the common procedure of cow’s milk pasteurization (75 °C for 15 s) does not lead to a significant destruction of MDE miR-155. Mutai et al. [446] detected miR-155-5p in bovine MDEs. Notably, the stability of miR-155 in raw cow’s milk was not affected by incubation at 37 °C for 1 h without treatment, incubation at 37 °C for 1 h with RNase (10 U/mL RNase A and 400 U/mL RNase H), exposure to a low pH (pH 2), or treatment with detergent (1% Triton X-100) compared to untreated bovine milk [503]. Remarkably, pretreatment of IEC-6 cells with bovine MDEs increased intracellular levels of miR-155 [504]. It has recently been shown that the primary oncogene of EBV, latent membrane protein 1 (LMP1), upregulates the expression of miR-155 in EBV+ B cell lymphoma cell lines and associated exosomes targeting the anti-apoptotic protein FOXO3A [505]. Notably, resistance to Bruton’s tyrosine kinase (BTK) inhibition by ibrutinib was associated with the downregulation of FOXO3A and PTEN levels and activation of AKT [506].
In fact, high levels of EV-associated miR-155 were found to correlate with chemotherapy resistance in several common cancers including DLBCL [507]. In patients with ABC DLBCL, Zare et al. [378] showed that exosomal miR-155 expression was upregulated in refractory/relapsed patients compared to responsive patients and patients receiving R-CHOP. Therefore overexpression of exosomal miR-155 in refractory/relapsed patients might be associated with more aggressive disease, poor response to R-CHOP therapy, and adverse prognosis [377]. In HCV-infected patients with rheumatoid arthritis, rituximab declined exosomal miR-155 concentrations [508]. Notably, a transfer of exosomal KSHV-miR-K12-11 (miR-K12-11) that has an identical seed sequence as miR-155, from KSHV-infected BCBL-1 and BC-1 lymphoma lines to T cells has been reported [509].
Recent evidence indicates that miR-155 enhanced lymphoma cell programmed death ligand 1 (PD-L1) expression, whereas PD-L1 blockade particularly retarded miR-155-overexpressing tumor growth consistent with the maintenance of CD8+ T cells and their function [510]. Targeting the polarization of tumor-associated macrophages and modulating miR-155 expression might be a new approach to treating DLBCL in the elderly [511].

4.3.7. MicroRNA-148a

miR-148a is a signature miR of commercial cow’s milk [447] and human milk and milk EVs [512,513]. In contrast to the boiling or UHT of cow’s milk, pasteurization and homogenization did not result in losses of miR-148a-3p compared to reeds of raw cow’s milk [514]. Benmoussa et al. [515] found that the bulk of bovine milk miRs including bta-miR-148a and bta-miR-125b sediment at 12,000 g and 35,000 g. Nevertheless, miR-148a is an abundant non-coding RNA detected in human [516,517], bovine [411], and porcine MDEs [518]. Human and bovine miR-148a-3p shares identical nucleotide sequences [519]. Recent evidence underlines that oral administration of exosomal bovine miR-148a-3p is bioavailable and reaches distant target tissues of C57BL6/mice [400]. Bovine MDE miR-148a-3p targets DNMT1 [427], the key maintenance DNA methyltransferase linking milk signaling to epigenetic regulation [519]. Guo et al. [517] recently demonstrated that human MDE miR-148a-3p directly targets TP53. Thus, MDEs via transfer of miR-148a-3p and miR-125b-5p to recipient cells may down-regulate the expression of p53 [520]. Of importance, p53 regulates the baseline expression of key genes involved in cell homeostasis such as FOXO1, PTEN, SESN1, SESN2, AR, IGF1R, BAK1, BIRC5, and TNFSF10 [521]. In fact, aberrations of the TP53 gene and dysregulation of the p53 pathway are regarded to be important in the pathogenesis of DLBCL [327]. Remarkably, TP53 loss led to an upregulation of programmed death ligand 1 (PD-L1) cell surface expression and secretion of EVs with EV-bound PD-L1 by lymphoma cells [522]. PTEN is a direct target of miR-148a-3p [523,524,525]. In fact, Reif et al. [526] demonstrated that human MDEs via miR-148a-3p down-regulated PTEN expression after incubation of normal fetal colon epithelial cells with MDEs. It is noteworthy that PTEN deficiency or PTEN loss are common adverse features in the pathogenesis of DLBCL [527,528,529,530].
BCL6 is a transcriptional repressor critically involved in the development and maintenance of GCs and lymphomagenesis [531]. Mutations and translocations leading to the sustained activity of BCL6 promote the development of GC-derived lymphomas [531]. Epigenetic mechanisms also lead to B cell hyperactivation [532,533]. In fact, BCL6 activity is modified by epigenetic mechanisms. As mentioned earlier, AICDA and DNMT1 form a complex to maintain methylation of the BCL6 promoter thereby inhibiting its expression. Loss of either AICDA or DNMT1 causes instability of the AICDA-DNMT1 complex resulting in its disassociation from the BCL6 promoter enhancing BCL6 expression in DLBCL [28] (Figure 2).
Importantly, DNMT1 is a direct target of MDE miR-148a-3p [235,526,534], whereas AICDA is targeted by miR-155-5p [535,536,537] and miR-29b [538]. Thus, continued consumption of pasteurized milk with a continuous transfer of MDE miR-148a, miR-155, and miR-29b via suppressing the AICDA-DNMT1 complex on the BCL6 promoter may maintain high expression of the oncogene BCL6 promoting lymphomagenesis.
Alles et al. [539] recently reported that miR-148a-3p impairs RAS/ERK signaling in B lymphocytes by targeting the Son of Sevenless 1 (SOS1) and SOS2 proteins. Notably, increased expression of miR-148a-3p reduced the expression of ERK1/ERK2 [539]. ERKs are essential for the differentiation of B cells into antibody-producing plasma cells and induce the expression of BLIMP1, a transcriptional repressor and “master regulator” of plasma cell differentiation [294,540,541,542]. In contrast, it has also been shown that miR-148a promotes plasma cell differentiation via the targeting of the BLIMP1 and IRF4 repressors MITF and BACH2 [384]. Premature expression of miR-148a by retroviral transduction favored plasmablast differentiation and the survival of in vitro activated primary murine B cells [384].
There is recent interest in PD-1/PD-L1 pathway blockade in patients with DLBCL [384,543,544,545,546,547,548,549]. Remarkably, Ashizawa et al. [550] demonstrated that PD-L1 is a direct target of miR-148a-3p in colorectal cancer cells. However, with the exception of the miR-148a/152 family, there is no other MDE-associated miR known to target PD-L1 [551]. Thus, miR-148a-3p may exert both tumor-promoting and protective effects in the pathogenesis of DLBCL (Table 3).

5. Discussion

Components of commercial cow’s milk activate mTORC1 signal transduction [53,54] and MDE miR-based epigenetic regulatory signaling [526], which share common pathways upregulated in DLBLC (Figure 1 and Figure 2). The transcriptional repressor BCL6 controls a large transcriptional network that is required for the formation and maintenance of GCs. GC B-cells represent the normal counterpart of most human B-cell lymphomas, which are often characterized by upregulated BCL6 expression and BCL6-mediated pathways [552]. BCL6 originally identified as encoded by a frequently translocated locus in DLBCLs [553], serves as a master regulator of the GC reaction [296,297]. BCL6 plays a central role in normal B cell development as well as lymphomagenesis [554]. Importantly, recent evidence indicates that BCL6 expression is regulated by epigenetic mechanisms involving the complex formation of AICDA and DNMT1 at the BCL6 promoter, which attenuates BCL6 expression through DNA methylation [28]. Human breastmilk and cow’s milk mediated transfer of MDE miRs that attenuate the expression of DNMT1 (miR-148a) and AICDA (miR-155, miR-29b) may thus enhance BCL6 expression promoting GC formation. This may represent a physiological effect on the newborn infant during the period of lactation. However, there is only limited information on the impact of breastmilk and its components on the epigenetic programming of immune function and immune development in early life [555]. Notably, BCL6 is critical for the development of a diverse primary B cell repertoire [556]. Importantly, there is intensive reciprocal and antagonistic crosstalk between BCL6 and BLIMP1 [557]. Lymphocytes with higher expression of BCL6 exhibit greater proliferative capacity but less secretory capacity, whereas lymphocytes with higher expression of BLIMP1 exhibit lower proliferative capacity and greater secretory capacity [556]. Of note, BLIMP1 is also regulated by miRs [321]. Especially, let-7 and miR-125b, which are overexpressed in DLBCL, suppress BLIMP1 expression [324,325,326]. BLIMP1 drives terminal differentiation in B cells and promotes plasmablast formation and antibody secretion [295]. The abundant milk-derived miRs of the let-7 family as well as miR-125b via targeting PRDM1, the gene encoding BLIMP1, may help to restrict unnecessary plasmablast and plasma cell formation and antibody synthesis during the postnatal period of lactation. During lactation, human milk provides abundant specific antibodies that protect the infant [558]. This is necessary because neonates exhibit an immature immune system and their immune activities are different from the activities of the adult immune system [559,560]. Remarkably, miRs are involved in the regulation of the immature neonatal immune system [560]. Neonatal and adult B cells are comparable in their proliferative responses to cooperative cell interactions. However, a marked deficiency in the ability of neonatal B cells to mature to immunoglobulin secretion was observed [561]. Apparently, “early” B lymphocytes are intrinsically defective in their ability to secrete immunoglobulin upon cooperative induction, whereas they show full competence to expand clonally [561]. Milk-derived miR signaling may epigenetically enhance the expression BCL6 expression and reduce the expression of BLIMP1 during the period of lactation thus promoting B lymphocyte proliferation and suppressing B cell differentiation and function. Weaning, the termination of breastmilk, maternal antibodies, and MDE miRs, may initiate an “epigenetic switch” attenuating BCL6 and enhancing BLIMP1 expression required for B cell differentiation to plasma cells and own antibody secretion by the infant [557]. This scenario is in accordance with the MDE miR-mediated switch of proliferating pancreatic β-cells to differentiated insulin-secreting matured β-cells after weaning [562,563]. Continued consumption of commercial cow’s milk via transfer of highly conserved MDE miRs may not only de-differentiate the pancreatic β-cell back to the neonatal phenotype but may also over-stimulate BCL6-driven B cell proliferation, a potential driving force in the pathogenesis of DLBCL (Figure 4).
Key new findings implicate DNA methylation heterogeneity as a core feature of DLBCL underlining the role of epigenetic dysfunction on lymphomagenesis [20,21,22,26,564,565,566,567]. Current findings highlight the potential role of miRs (lymphomiRs) as important factors in lymphomagenesis [568]. At present, it is not possible to oversee the potential contribution of bovine MDE miRs and their targets in epigenomic dysregulations of DLBCL and its subtypes (ABC, GCB) during its various stages of lymphomagenesis. Nevertheless, the presented epidemiological and mechanistic insights link the consumption of cow’s milk and bovine MDE miRs to the pathogenesis of DLBCL.
It is noteworthy that the distribution and consumption of pasteurized cow’s milk is a new human behavior in relation to the human history of milk consumption since ancient times [387]. MDEs and MDE miRs survive pasteurization, whereas MDE and milk miRs are destroyed by boiling or UHT [389,390] and are attacked by fermentation [448]. Thus, future epidemiological studies relating to milk consumption and DLBCL risk have to consider the methods of thermal milk processing. Whereas MDE miRs may function as a pathogenic factor in DLBCL, MDE miRs may exert beneficial effects in the prevention of colorectal cancer (CRC). The consumption of cow’s milk has been related to a decreased risk of CRC [569,570,571,572]. In fact, bovine MDEs exhibited anti-inflammatory effects in murine models of colitis [573,574]. Notably, miR-148a-3p and MDE miR-27b exert tumor suppressive functions in CRC [575,576,577].

6. Conclusions

Whereas MDEs and their miRs are of key importance for neonatal development [578,579], MDEs may exert pathogenic effects in adults [579]. In this light, bovine MDEs should be regarded as pathogens that have to be excluded from the human food chain [580,581]. With regard to DLBCL pathogenesis, we are concerned about recent recommendations promoting bovine miR-155-rich colostrum for “immune-nutrition” in elderly subjects [582] as well as bovine MDEs as “health-improving bioactive ingredients” in the context of human nutrition [583]. Persistent exposure to bovine miR-155-rich colostral MDEs may thus function like EBV-driven exosomal overexpression of miR-155 inducing lymphomagenesis [505]. In addition, medical advice to include whole cow’s milk in the diet for patients with DLBCL is questionable [584]. Obviously, there is an urgent need to study milk as a biological system [585], which is the basis to understand milk’s beneficial and adverse effects on human health during the differing stages of immune system development in infancy and homeostatic maintenance of immune functions in adulthood [586]. DLBCL accounts for approximately 30% of adult lymphomas and 10% of lymphomas diagnosed before the age of 18 years [587]. Importantly, the incidence of DLBCL increases with age [588]. The average age at the initial diagnosis of DLBCL is approximately 70 years [589,590]. Unfortunately, current epidemiological studies do not report lifetime periods of cow’s milk exposure in relation to the occurrence of DLBCL. Thus, there is no information on potential vulnerable windows for cow’s milk consumption during infancy or adolescence and the later onset of DLBCL as demonstrated for prostate cancer [591]. The dietary exposure of humans to MDEs and miRs beyond the weaning period is an unnatural intervention that may represent a pathogenic risk factor for all age groups [52]. Cow’s milk-mediated transmission of oncogenic viruses is a recent matter of concern that requires further investigation. In contrast to cow’s milk and dairy products, a higher intake of green leafy vegetables and cruciferous vegetables has been associated with a lower overall risk of NHL, particularly DLBCL and follicular lymphoma [48].

7. Limitations

We would like to note that the majority of reported associations between milk signaling and pathogenic signaling in DLBCL are based on literature research data and mechanistic models not on direct experimental evidence. Data that clarify the potential impact of MDE miRs on B cells as well as B cell lymphoma cells are still missing and need to be explored in experimental settings and studies including human milk consumers. The recently presented epidemiological evidence linking cow’s milk consumption to the risk of DLBCL also requires more detailed data on thermal milk processing (destruction of MDE miRs by UHT or bioavailability of MDE miRs in pasteurized milk). Because only a smaller fraction of human milk consumers will develop DLBCL during their lifetime, predisposing individual factors on the patient’s side may be a greater importance. Nevertheless, milk signaling evidently aggravates signal transduction pathways and epigenetic deviations contributing to DLBCL initiation and progression. Data on epigenetic regulatory effects of breastmilk signaling in healthy newborn infants and potential epigenetics shifts after weaning are missing and appear to be of utmost importance to understanding the physiological impact of milk signaling on postnatal B cell proliferation and development. Future studies that determine the epigenetic heterogeneity in human milk consumers versus individuals who were not exposed to cow’s milk (lactose intolerance) may provide deeper insights into the milk-DLBCL relationship.

8. Materials and Methods

We conducted our bibliographic research exclusively via PubMed [https://pubmed.ncbi.nlm.nih.gov] between 1995–2022 using various keywords such as “B cell”, “B lymphocyte”, ”B cell proliferation“, B cell differentiation”, “malignant B cell”, “malignant B lymphocyte”, “diffuse large B cell lymphoma”, “DLBCL”, “non-Hodgkin lymphoma”, “lymphomagenesis”, lymphocyte-induced maturation protein 1”, “BLIMP1”, “PR domain-containing protein 1”, “PRDM1”, “B cell lymphoma 6”, “BCL6”, “milk”, “cow’s milk”, “dairy”, “milk exosome”, “extracellular vesicle”, “EV”, “microRNA”, “miRNA”, “miR”, “exosomal miR”, “epigenetic”, and “epigenetic regulation”. We also analyzed papers based on the bibliographic references cited by the studies found on PubMed during our search. Whenever possible, we selected the most recent and comprehensive reviews on the topic in question. All selected articles were written in English.

Author Contributions

Writing—original draft preparation, B.C.M.; writing—review and editing, B.C.M., R.S., R.W., C.L. and G.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ABCactivated B-cell like
AICDAactivation-induced cytidine deaminase
AMPKAMP-activated protein kinase
ATF4activating transcription factor 4
BACH2BTB and CNC homology 2
BCAAbranched-chain amino acid
BCL6B cell lymphoma 6
BCL10B cell CLL/lymphoma 10
BCL11BB cell lymphoma/leukemia 11B
BCRB cell receptor
BICB cell integration cluster
BIMBCL2-like 11
BIRC5baculoviral IAP repeat-containing protein 5
BLIMP1B cell differentiation B lymphocyte-induced maturation protein 1
BLVbovine leukemia virus
BMMFbovine meat and milk factor
BPAbisphenol A
CARD11caspase recruitment domain-containing protein 11
CDKN1Acyclin-dependent kinase inhibitor 1A
C-MYKMYC protooncogene
CRESScircular replicase-encoding single-stranded
CTNNB1catenin β1
DEPTORDEP domain-containing mTOR-interacting protein
DLBCLdiffuse large B cell lymphoma
DNMT1DNA methyltransferase 1
EBNA2EBV nuclear antigen 2
E1estrone
E2estradiol
E3estriol
EBVEpstein-Barr virus
E2F1E2F transcription factor 1
eIF4Beukaryotic translation initiation factor 4B
eIF4Eeukaryotic translation initiation factor 4E
ERβestrogen receptor-β
EVextracellular vesicle
FcRnneonatal Fc receptor
FoxO1Aforkhead box O1A
FoxO3Aforkhead box O3A
GADD45ADNA-damage-inducible gene α
GCgerminal center
GCBgerminal center B-cell like
GHgrowth hormone
GHRgrowth hormone receptor
GPR30G protein-coupled estrogen receptor 1
HBVhepatitis B virus
HCVhepatitis C virus
HHV8human herpes virus 8
HIF-1hypoxia-inducible factor 1
HIVhuman immunodeficiency virus
HPVhuman papilloma virus
HTLV1human T-cell leukemia virus type 1
IGF-1insulin-like growth factor 1
IGF1Rinsulin-like growth factor 1 receptor
IGFPB3insulin-like growth factor binding protein 3
IKKinhibitor of kB kinase
INPP5inositol polyphosphate-5-phosphatase, 145-KD (SHIP1)
IRF4interferon regulatory factor 4
KSHVKaposi sarcoma-associated herpesvirus
LAT1L-type neutral amino acid transporter 1
MALT1mucosa-associated lymphoid tissue lymphoma translocation gene 1
MAXMAX protein
MDEmilk-derived exosome
MDM2MDM2 protooncogene
MDR1ATP-binding cassette, subfamily B, member 1 (ABCB1)
MDSCmyeloid-derived suppressor cells
miRmicro-ribonucleic acid
MITFmicrophthalmia-associated transcription factor
MSCmesenchymal stem cell
MYCMYC protooncogene, bHLH transcription factor
MYD88MYD88 innate immune signal transduction adaptor
mTORC1mechanistic target of rapamycin complex 1
MXD1MAX dimerization protein 1
MXD4MAX dimerization protein 4
NF-κBnuclear factor kappa B
NHLnon-Hodgkin lymphoma
PBMCperipheral blood mononuclear cell
PDCD4programmed cell death 4
PDK1phosphoinositide-dependent kinase 1
PI3Kphosphatidylinositol-3 kinase
PIK3IP1phosphatidylinositol 3-kinase-interacting protein 1
PIK3R1PI3K regulatory subunit 1
PPPpicropodophyllin
PRDM1PR domain-containing protein 1 (BLIMP1)
PTENphosphatase and tensin homolog
RAGRagulator
R-CHOPrituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone regimen
RISCRNA-induced silencing complex
RUNX2runt-related transcription factor 2
SIRT3sirtuin 3
SMOSmoothened
SOSSon of Sevenless
STAT3signal transducer and activator of transcriptin 3
SV40Simian virus 40
TAMtumor-associated macrophage
TCAtricarboxylic acid
TCL1T-cell leukemia gene 1
TLRToll-like receptor
TMEtumor microenvironment
TNFAIP3tumor necrosis factor-α induced protein 3
TNFRSF10Btumor necrosis factor receptor superfamily, member 10B
TP53tumor protein p53
TRAF2TNF receptor-associated factor 2
TSC2tuberin
TWIST1TWIST family bHLH transcription factor 1
UHTultraheat treatment
UTRuntranslated region
VHLvon Hippel–Lindau tumor suppressor
YAPHippo-Yes-associated protein

References

  1. Martelli, M.; Ferreri, A.J.; Agostinelli, C.; Di Rocco, A.; Pfreundschuh, M.; Pileri, S.A. Diffuse large B-cell lymphoma. Crit. Rev. Oncol. Hematol. 2013, 87, 146–171. [Google Scholar] [PubMed]
  2. Li, S.; Young, K.H.; Medeiros, L.J. Diffuse large B-cell lymphoma. Pathology 2018, 50, 74–87. [Google Scholar] [PubMed] [Green Version]
  3. Wilcox, R.A. Cutaneous B-cell lymphomas: 2019 update on diagnosis, risk stratification, and management. Am. J. Hematol. 2018, 93, 1427–1430. [Google Scholar] [PubMed] [Green Version]
  4. Stadler, R.; Stranzenbach, R. Molecular pathogenesis of cutaneous lymphomas. Exp. Dermatol. 2018, 27, 1078–1083. [Google Scholar] [PubMed] [Green Version]
  5. Dippel, E.; Assaf, C.; Becker, J.C.; von Bergwelt-Baildon, M.; Bernreiter, S.; Cozzio, A.; Eich, H.T.; Elsayad, K.; Follmann, M.; Grabbe, S.; et al. S2k-Guidelines-Cutaneous lymphomas (ICD10 C82–C86): Update 2021. J. Dtsch. Dermatol. Ges. 2022, 20, 537–554. [Google Scholar]
  6. Chihara, D.; Johnston, K.; Bolatova, T.; Szabo, S.; Kalsekar, A.; Mutebi, A.; Yang, H.; Liu, Y.; Attinson, D.; Hutchings, M. An epidemiological model to estimate the prevalence of diffuse large B-cell lymphoma in the United States. Clin. Lymphoma Myeloma Leuk. 2022, 22, e1092–e1099. [Google Scholar]
  7. Kanas, G.; Ge, W.; Quek, R.G.W.; Keeven, K.; Nersesyan, K.; Arnason, J.E. Epidemiology of diffuse large B-cell lymphoma (DLBCL) and follicular lymphoma (FL) in the United States and Western Europe: Population-level projections for 2020–2025. Leuk. Lymphoma 2022, 63, 54–63. [Google Scholar]
  8. Alizadeh, A.A.; Eisen, M.B.; Davis, R.E.; Ma, C.; Lossos, I.S.; Rosenwald, A.; Boldrick, J.C.; Sabet, H.; Tran, T.; Yu, X.; et al. Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature 2000, 403, 503–511. [Google Scholar]
  9. Caro, P.; Kishan, A.U.; Norberg, E.; Stanley, I.A.; Chapuy, B.; Ficarro, S.B.; Polak, K.; Tondera, D.; Gounarides, J.; Yin, H.; et al. Metabolic signatures uncover distinct targets in molecular subsets of diffuse large B cell lymphoma. Cancer Cell 2012, 22, 547–560. [Google Scholar]
  10. Miao, Y.; Medeiros, L.J.; Li, Y.; Li, J.; Young, K.H. Genetic alterations and their clinical implications in DLBCL. Nat. Rev. Clin. Oncol. 2019, 16, 634–652. [Google Scholar]
  11. Liu, Y.; Barta, S.K. Diffuse large B-cell lymphoma: 2019 update on diagnosis, risk stratification, and treatment. Am. J. Hematol. 2019, 94, 604–616. [Google Scholar] [CrossRef] [Green Version]
  12. Morin, R.D.; Mungall, K.; Pleasance, E.; Mungall, A.J.; Goya, R.; Huff, R.D.; Scott, D.W.; Ding, J.; Roth, A.; Chiu, R.; et al. Mutational and structural analysis of diffuse large B-cell lymphoma using whole-genome sequencing. Blood 2013, 122, 1256–1265. [Google Scholar] [CrossRef] [Green Version]
  13. Ando, M.; Sato, Y.; Takata, K.; Nomoto, J.; Nakamura, S.; Ohshima, K.; Takeuchi, T.; Orita, Y.; Kobayashi, Y.; Yoshino, T. A20 (TNFAIP3) deletion in Epstein-Barr virus-associated lymphoproliferative disorders/lymphomas. PLoS ONE 2013, 8, e56741. [Google Scholar]
  14. Davis, R.E.; Ngo, V.N.; Lenz, G.; Tolar, P.; Young, R.M.; Romesser, P.B.; Kohlhammer, H.; Lamy, L.; Zhao, H.; Yang, Y.; et al. Chronic active B-cell-receptor signalling in diffuse large B-cell lymphoma. Nature 2010, 463, 88–92. [Google Scholar]
  15. Ngo, V.N.; Young, R.M.; Schmitz, R.; Jhavar, S.; Xiao, W.; Lim, K.H.; Kohlhammer, H.; Xu, W.; Yang, Y.; Zhao, H.; et al. Oncogenically active MYD88 mutations in human lymphoma. Nature 2011, 470, 115–119. [Google Scholar]
  16. Pasqualucci, L.; Dominguez-Sola, D.; Chiarenza, A.; Fabbri, G.; Grunn, A.; Trifonov, V.; Kasper, L.H.; Lerach, S.; Tang, H.; Ma, J.; et al. Inactivating mutations of acetyltransferase genes in B-cell lymphoma. Nature 2011, 471, 189–195. [Google Scholar]
  17. Zhang, J.; Grubor, V.; Love, C.L.; Banerjee, A.; Richards, K.L.; Mieczkowski, P.A.; Dunphy, C.; Choi, W.; Au, W.Y.; Srivastava, G.; et al. Genetic heterogeneity of diffuse large B-cell lymphoma. Proc. Natl. Acad. Sci. USA 2013, 110, 1398–1403. [Google Scholar]
  18. Pasqualucci, L.; Dalla-Favera, R. Genetics of diffuse large B-cell lymphoma. Blood 2018, 131, 2307–2319. [Google Scholar]
  19. Schmitz, R.; Wright, G.W.; Huang, D.W.; Johnson, C.A.; Phelan, J.D.; Wang, J.Q.; Roulland, S.; Kasbekar, M.; Young, R.M.; Shaffer, A.L.; et al. Genetics and pathogenesis of diffuse large B-cell lymphoma. N. Engl. J. Med. 2018, 378, 1396–1407. [Google Scholar] [CrossRef]
  20. De, S.; Shaknovich, R.; Elemento, O.; Geng, H.; Kormaksson, M.; Jiang, Y.; Woolcock, B.; Johnson, N.; Polo, J.M. Aberration in DNA methylation in B-cell lymphomas has a complex origin and increases with disease severity. PLoS Genet. 2013, 9, e1003137. [Google Scholar] [CrossRef] [Green Version]
  21. Jiang, Y.; Melnick, A. The epigenetic basis of diffuse large B-cell lymphoma. Semin. Hematol. 2015, 52, 86–96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Jiang, Y.; Dominguez, P.M.; Melnick, A.M. The many layers of epigenetic dysfunction in B-cell lymphomas. Curr. Opin. Hematol. 2016, 23, 377–384. [Google Scholar] [CrossRef] [PubMed]
  23. Frazzi, R.; Zanetti, E.; Pistoni, M.; Tamagnini, I.; Valli, R.; Braglia, L.; Merli, F. Methylation changes of SIRT1, KLF4, DAPK1 and SPG20 in B-lymphocytes derived from follicular and diffuse large B-cell lymphoma. Leuk. Res. 2017, 57, 89–96. [Google Scholar] [CrossRef] [PubMed]
  24. Bakhshi, T.J.; Georgel, P.T. Genetic and epigenetic determinants of diffuse large B-cell lymphoma. Blood Cancer J. 2020, 10, 123. [Google Scholar]
  25. Oricchio, E. Epigenetic balance in DLBCL. Blood 2021, 138, 355–356. [Google Scholar] [CrossRef]
  26. Isshiki, Y.; Melnick, A. Epigenetic mechanisms of therapy resistance in diffuse large B cell lymphoma (DLBCL). Curr. Cancer Drug Targets. 2021, 21, 274–282. [Google Scholar] [CrossRef]
  27. Heward, J.; Konali, L.; D’Avola, A.; Close, K.; Yeomans, A.; Philpott, M.; Dunford, J.; Rahim, T.; Al Seraihi, A.F.; Wang, J.; et al. KDM5 inhibition offers a novel therapeutic strategy for the treatment of KMT2D mutant lymphomas. Blood 2021, 138, 370–381. [Google Scholar] [CrossRef]
  28. Jiao, J.; Lv, Z.; Zhang, P.; Wang, Y.; Yuan, M.; Yu, X.; Otieno Odhiambo, W.; Zheng, M.; Zhang, H.; Ma, Y.; et al. AID assists DNMT1 to attenuate BCL6 expression through DNA methylation in diffuse large B-cell lymphoma cell lines. Neoplasia 2020, 22, 142–153. [Google Scholar] [CrossRef]
  29. Shen, H.M.; Peters, A.; Baron, B.; Zhu, X.; Storb, U. Mutation of BCL-6 gene in normal B cells by the process of somatic hypermutation of Ig genes. Science 1998, 280, 1750–1752. [Google Scholar] [CrossRef]
  30. Thandra, K.C.; Barsouk, A.; Saginala, K.; Padala, S.A.; Barsouk, A.; Rawla, P. Epidemiology of non-Hodgkin’s lymphoma. Med. Sci. 2021, 9, 5. [Google Scholar] [CrossRef]
  31. Zhang, Y.; Sanjose, S.D.; Bracci, P.M.; Morton, L.M.; Wang, R.; Brennan, P.; Hartge, P.; Boffetta, P.; Becker, N.; Maynadie, M.; et al. Personal use of hair dye and the risk of certain subtypes of non-Hodgkin lymphoma. Am. J. Epidemiol. 2008, 167, 1321–1331. [Google Scholar] [CrossRef] [Green Version]
  32. Guo, H.; Bassig, B.A.; Lan, Q.; Zhu, Y.; Zhang, Y.; Holford, T.R.; Leaderer, B.; Boyle, P.; Qin, Q.; Zhu, C.; et al. Polymorphisms in DNA repair genes, hair dye use, and the risk of non-Hodgkin lymphoma. Cancer Causes Control 2014, 25, 1261–1270. [Google Scholar] [CrossRef] [Green Version]
  33. Kleinstern, G.; Abu Seir, R.; Perlman, R.; Khatib, A.; Abdeen, Z.; Elyan, H.; Nirel, R.; Amir, G.; Ramlawi, A.; Sabatin, F.; et al. Ethnic variation in medical and lifestyle risk factors for B cell non-Hodgkin lymphoma: A case-control study among Israelis and Palestinians. PLoS ONE 2017, 12, e0171709. [Google Scholar] [CrossRef] [Green Version]
  34. Eriksson, M.; Hardell, L.; Carlberg, M.; Akerman, M. Pesticide exposure as risk factor for non-Hodgkin lymphoma including histopathological subgroup analysis. Int. J. Cancer 2008, 123, 1657–1663. [Google Scholar] [CrossRef]
  35. Boffetta, P.; Ciocan, C.; Zunarelli, C.; Pira, E. Exposure to glyphosate and risk of non-Hodgkin lymphoma: An updated meta-analysis. Med. Lav. 2021, 112, 119–194. [Google Scholar]
  36. Pahwa, M.; Beane Freeman, L.E.; Spinelli, J.J.; Blair, A.; McLaughlin, J.R.; Zahm, S.H.; Cantor, K.P.; Weisenburger, D.D.; Punam Pahwa, P.P.; Dosman, J.A.; et al. Glyphosate use and associations with non-Hodgkin lymphoma major histological sub-types: Findings from the North American Pooled Project. Scand. J. Work Environ. Health 2019, 45, 600–609. [Google Scholar] [CrossRef] [Green Version]
  37. Chen, Y.K.; Tan, Y.Y.; Yao, M.; Lin, H.C.; Tsai, M.H.; Li, Y.Y.; Hsu, Y.J.; Huang, T.T.; Chang, C.W.; Cheng, C.M.; et al. Bisphenol A-induced DNA damages promote to lymphoma progression in human lymphoblastoid cells through aberrant CTNNB1 signaling pathway. iScience 2021, 24, 102888. [Google Scholar] [CrossRef]
  38. Sapkota, S.; Shaikh, H. Non-Hodgkin lymphoma. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2022. [Google Scholar]
  39. Franceschi, S.; Serraino, D.; Carbone, A.; Talamini, R.; La Vecchia, C. Dietary factors and non-Hodgkin’s lymphoma: A case-control study in the northeastern part of Italy. Nutr. Cancer 1989, 12, 333–341. [Google Scholar] [CrossRef]
  40. Ward, M.H.; Zahm, S.H.; Weisenburger, D.D.; Gridley, G.; Cantor, K.P.; Saal, R.C.; Blair, A. Dietary factors and non-Hodgkin’s lymphoma in Nebraska (United States). Cancer Causes Control 1994, 5, 422–432. [Google Scholar]
  41. Chang, E.T.; Smedby, K.E.; Zhang, S.M.; Hjalgrim, H.; Melbye, M.; Ost, A.; Glimelius, B.; Wolk, A.; Adami, H.O. Dietary factors and risk of non-Hodgkin lymphoma in men and women. Cancer Epidemiol. Biomark. Prev. 2005, 14, 512–520. [Google Scholar] [CrossRef] [Green Version]
  42. Skibola, C.F. Obesity, diet and risk of non-Hodgkin lymphoma. Cancer Epidemiol. Biomark. Prev. 2007, 16, 392–395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Balasubramaniam, G.; Saoba, S.; Sarade, M.; Pinjare, S. Case-control study of risk factors for non-Hodgkin lymphoma in Mumbai, India. Asian Pac. J. Cancer Prev. 2013, 14, 775–780. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Chiu, B.C.; Cerhan, J.R.; Folsom, A.R.; Sellers, T.A.; Kushi, L.H.; Wallace, R.B.; Zheng, W.; Potter, J.D. Diet and risk of non-Hodgkin lymphoma in older women. JAMA 1996, 275, 1315–1321. [Google Scholar] [CrossRef] [PubMed]
  45. Zhang, S.M.; Hunter, D.J.; Rosner, B.A.; Giovannucci, E.L.; Colditz, G.A.; Speizer, F.E.; Willett, W.C. Intakes of fruits, vegetables, and related nutrients and the risk of non-Hodgkin’s lymphoma among women. Cancer Epidemiol. Biomark. Prev. 2000, 9, 477–485. [Google Scholar]
  46. Thompson, C.A.; Habermann, T.M.; Wang, A.H.; Vierkant, R.A.; Folsom, A.R.; Ross, J.A.; Cerhan, J.R. Antioxidant intake from fruits, vegetables and other sources and risk of non-Hodgkin’s lymphoma: The Iowa Women’s Health Study. Int. J. Cancer 2010, 126, 992–1003. [Google Scholar] [CrossRef]
  47. Ali, A.; Al-Belushi, B.S.; Waly, M.I.; Al-Moundhri, M.; Burney, I.A. Dietary and lifestyle factors and risk of non-Hodgkin’s lymphoma in Oman. Asian Pac. J. Cancer Prev. 2013, 14, 841–848. [Google Scholar] [CrossRef] [Green Version]
  48. Chiu, B.C.; Kwon, S.; Evens, A.M.; Surawicz, T.; Smith, S.M.; Weisenburger, D.D. Dietary intake of fruit and vegetables and risk of non-Hodgkin lymphoma. Cancer Causes Control 2011, 22, 1183–1195. [Google Scholar] [CrossRef]
  49. Wang, J.; Li, X.; Zhang, D. Dairy product consumption and risk of non-Hodgkin lymphoma: A meta-analysis. Nutrients 2016, 8, 120. [Google Scholar] [CrossRef] [Green Version]
  50. Kakkoura, M.G.; Du, H.; Guo, Y.; Yu, C.; Yang, L.; Pei, P.; Chen, Y.; Sansome, S.; Chan, W.C.; Yang, X.; et al. China Kadoorie Biobank (CKB) Collaborative Group. Dairy consumption and risks of total and site-specific cancers in Chinese adults: An 11-year prospective study of 0.5 million people. BMC Med. 2022, 20, 134. [Google Scholar] [CrossRef]
  51. Saberi Hosnijeh, F.; Casabonne, D.; Nieters, A.; Solans, M.; Naudin, S.; Ferrari, P.; Mckay, J.D.; Benavente, Y.; Weiderpass, E.; Freisling, H.; et al. Association between anthropometry and lifestyle factors and risk of B-cell lymphoma: An exposome-wide analysis. Int. J. Cancer 2021, 148, 2115–2128. [Google Scholar] [CrossRef]
  52. Melnik, B.C. Lifetime impact of cow’s milk on overactivation of mTORC1: From fetal to childhood overgrowth, acne, diabetes, cancers, and neurodegeneration. Biomolecules 2021, 11, 404. [Google Scholar] [CrossRef]
  53. Melnik, B.C.; John, S.M.; Schmitz, G. Milk is not just food but most likely a genetic transfection system activating mTORC1 signaling for postnatal growth. Nutr. J. 2013, 12, 103. [Google Scholar]
  54. Melnik, B.C. Milk—A nutrient system of mammalian evolution promoting mTORC1-dependent translation. Int. J. Mol. Sci. 2015, 16, 17048–17087. [Google Scholar] [CrossRef]
  55. Sergentanis, T.N.; Ntanasis-Stathopoulos, I.; Tzanninis, I.G.; Gavriatopoulou, M.; Sergentanis, I.N.; Dimopoulos, M.A.; Psaltopoulou, T. Meat, fish, dairy products and risk of hematological malignancies in adults—A systematic review and meta-analysis of prospective studies. Leuk. Lymphoma 2019, 60, 1978–1990. [Google Scholar] [CrossRef]
  56. Iso, H.; Kubota, Y.; Japan Collaborative Cohort Study for Evaluation of Cancer. Nutrition and disease in the Japan Collaborative Cohort Study for Evaluation of Cancer (JACC). Asian Pac. J. Cancer Prev. 2007, 8, 35–80. [Google Scholar]
  57. Rohrmann, S.; Linseisen, J.; Jakobsen, M.U.; Overvad, K.; Raaschou-Nielsen, O.; Tjonneland, A.; Boutron-Ruault, M.C.; Kaaks, R.; Becker, N.; Bergmann, M.; et al. Consumption of meat and dairy and lymphoma risk in the European Prospective Investigation into Cancer and Nutrition. Int. J. Cancer 2011, 128, 623–634. [Google Scholar] [CrossRef]
  58. Nwabo Kamdje, A.H.; Seke Etet, P.F.; Kipanyula, M.J.; Vecchio, L.; Tagne Simo, R.; Njamnshi, A.K.; Lukong, K.E.; Mimche, P.N. Insulin-like growth factor-1 signaling in the tumor microenvironment: Carcinogenesis, cancer drug resistance, and therapeutic potential. Front. Endocrinol. 2022, 13, 927390. [Google Scholar] [CrossRef]
  59. Kasprzak, A.; Kwasniewski, W.; Adamek, A.; Gozdzicka-Jozefiak, A. Insulin-like growth factor (IGF) axis in cancerogenesis. Mutat. Res. Rev. Mutat. Res. 2017, 772, 78–104. [Google Scholar]
  60. Hanahan, D.; Weinberg, R.A. The hallmarks of cancer. Cell 2000, 100, 57–70. [Google Scholar]
  61. Strömberg, T.; Feng, X.; Delforoush, M.; Berglund, M.; Lin, Y.; Axelson, M.; Larsson, O.; Georgii-Hemming, P.; Lennartsson, J.; Enblad, G. Picropodophyllin inhibits proliferation and survival of diffuse large B-cell lymphoma cells. Med. Oncol. 2015, 32, 188. [Google Scholar] [CrossRef]
  62. Ravindra, V.M.; Raheja, A.; Corn, H.; Driscoll, M.; Welt, C.; Simmons, D.L.; Couldwell, W.T. Primary pituitary diffuse large B-cell lymphoma with somatotroph hyperplasia and acromegaly: Case report. J. Neurosurg. 2017, 126, 1725–1730. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Zhou, X.; Zhang, Y.; Li, Y.; Xu, Y.; Zhang, L.; Li, Y.; Wang, X. Klotho suppresses tumor progression via inhibiting IGF-1R signaling in T-cell lymphoma. Oncol. Rep. 2017, 38, 967–974. [Google Scholar] [PubMed] [Green Version]
  64. Zhou, X.; Fang, X.; Jiang, Y.; Geng, L.; Li, X.; Li, Y.; Lu, K.; Li, P.; Lv, X.; Wang, X. Klotho, an anti-aging gene, acts as a tumor suppressor and inhibitor of IGF-1R signaling in diffuse large B cell lymphoma. J. Hematol. Oncol. 2017, 10, 37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Zhou, X.; Chen, N.; Xu, H.; Zhou, X.; Wang, J.; Fang, X.; Zhang, Y.; Li, Y.; Yang, J.; Wang, X. Regulation of Hippo-YAP signaling by insulin-like growth factor-1 receptor in the tumorigenesis of diffuse large B-cell lymphoma. J. Hematol. Oncol. 2020, 13, 77. [Google Scholar] [PubMed]
  66. Agarwal, N.K.; Kim, C.H.; Kunkalla, K.; Vaghefi, A.; Sanchez, S.; Manuel, S.; Bilbao, D.; Vega, F.; Landgraf, R. Smoothened (SMO) regulates insulin-like growth factor 1 receptor (IGF1R) levels and protein kinase B (AKT) localization and signaling. Lab. Investig. 2022, 102, 401–410. [Google Scholar] [CrossRef]
  67. Qin, L.Q.; He, K.; Xu, J.Y. Milk consumption and circulating insulin-like growth factor-I level: A systematic literature review. Int. J. Food Sci. Nutr. 2009, 60 (Suppl. S7), 330–340. [Google Scholar]
  68. Rich-Edwards, J.W.; Ganmaa, D.; Pollak, M.N.; Nakamoto, E.K.; Kleinman, K.; Tserendolgor, U.; Willett, W.C.; Frazier, A.L. Milk consumption and the prepubertal somatotropic axis. Nutr. J. 2007, 6, 28. [Google Scholar]
  69. Hoppe, C.; Mølgaard, C.; Juul, A.; Michaelsen, K.F. High intakes of skimmed milk, but not meat, increase serum IGF-I and IGFBP-3 in eight-year-old boys. Eur. J. Clin. Nutr. 2004, 58, 1211–1216. [Google Scholar]
  70. Hoppe, C.; Mølgaard, C.; Dalum, C.; Vaag, A.; Michaelsen, K.F. Differential effects of casein versus whey on fasting plasma levels of insulin, IGF-1 and IGF-1/IGFBP-3: Results from a randomized 7-day supplementation study in prepubertal boys. Eur. J. Clin. Nutr. 2009, 63, 1076–1083. [Google Scholar] [CrossRef] [Green Version]
  71. Barrea, L.; Di Somma, C.; Macchia, P.E.; Falco, A.; Savanelli, M.C.; Orio, F.; Colao, A.; Savastano, S. Influence of nutrition on somatotropic axis: Milk consumption in adult individuals with moderate-severe obesity. Clin. Nutr. 2017, 36, 293–301. [Google Scholar]
  72. Rogers, I.; Emmett, P.; Gunnell, D.; Dunger, D.; Holly, J.; ALSPAC Study Tteam. Milk as a food for growth? The insulin-like growth factors link. Public Health Nutr. 2006, 9, 359–368. [Google Scholar] [CrossRef] [Green Version]
  73. Norat, T.; Dossus, L.; Rinaldi, S.; Overvad, K.; Grønbaek, H.; Tjønneland, A.; Olsen, A.; Clavel-Chapelon, F.; Boutron-Ruault, M.C.; Boeing, H.; et al. Diet, serum insulin-like growth factor-I and IGF-binding protein-3 in European women. Eur. J. Clin. Nutr. 2007, 61, 91–98. [Google Scholar]
  74. Crowe, F.L.; Key, T.J.; Allen, N.E.; Appleby, P.N.; Roddam, A.; Overvad, K.; Grønbaek, H.; Tjønneland, A.; Halkjaer, J.; Dossus, L.; et al. The association between diet and serum concentrations of IGF-I, IGFBP-1, IGFBP-2, and IGFBP-3 in the European Prospective Investigation into Cancer and Nutrition. Cancer Epidemiol. Biomark. Prev. 2009, 18, 1333–1340. [Google Scholar] [CrossRef] [Green Version]
  75. Romo Ventura, E.; Konigorski, S.; Rohrmann, S.; Schneider, H.; Stalla, G.K.; Pischon, T.; Linseisen, J.; Nimptsch, K. Association of dietary intake of milk and dairy products with blood concentrations of insulin-like growth factor 1 (IGF-1) in Bavarian adults. Eur. J. Nutr. 2020, 59, 1413–1420. [Google Scholar] [CrossRef]
  76. Lovell, A.L.; Milne, T.; Matsuyama, M.; Hill, R.J.; Davies, P.S.W.; Grant, C.C.; Wall, C.R. Protein intake, IGF-1 concentrations, and growth in the second year of life in children receiving Growing Up Milk—Lite (GUMLi) or cow’s milk (CM) intervention. Front. Nutr. 2021, 8, 666228. [Google Scholar] [CrossRef]
  77. Honegger, A.; Humbel, R.E. Insulin-like growth factors I and II in fetal and adult bovine serum. Purification, primary structures, and immunological cross-reactivities. J. Biol. Chem. 1986, 261, 569–575. [Google Scholar]
  78. Wheelhouse, N.M.; Stubbs, A.K.; Lomax, M.A.; MacRae, J.C.; Hazlerigg, D.G. Growth hormone and amino acid supply interact synergistically to control insulin-like growth factor-I production and gene expression in cultured ovine hepatocytes. J. Endocrinol. 1999, 163, 353–361. [Google Scholar] [CrossRef] [Green Version]
  79. Stubbs, A.K.; Wheelhouse, N.M.; Lomax, M.A.; Hazlerigg, D.G. Nutrient-hormone interaction in the ovine liver: Methionine supply selectively modulates growth hormone-induced IGF-I gene expression. J. Endocrinol. 2002, 174, 335–341. [Google Scholar]
  80. Tsugawa, Y.; Handa, H.; Imai, T. Arginine induces IGF-1 secretion from the endoplasmic reticulum. Biochem. Biophys. Res. Commun. 2019, 514, 1128–1132. [Google Scholar] [CrossRef]
  81. Wan, X.; Wang, S.; Xu, J.; Zhuang, L.; Xing, K.; Zhang, M.; Zhu, X.; Wang, L.; Gao, P.; Xi, Q.; et al. Dietary protein-induced hepatic IGF-1 secretion mediated by PPARγ activation. PLoS ONE 2017, 12, e0173174. [Google Scholar] [CrossRef] [Green Version]
  82. Melnik, B.C.; Schmitz, G.; John, S.M. Health risks related to milk consumption: A critical evaluation from the medical perspective. MMW Fortschr. Med. 2021, 163 (Suppl. S4), 3–9. [Google Scholar] [CrossRef] [PubMed]
  83. Bounous, G.; Kongshavn, P.A. Differential effect of dietary protein type on the B-cell and T-cell immune responses in mice. J. Nutr. 1985, 115, 1403–1408. [Google Scholar] [CrossRef] [PubMed]
  84. Bounous, G.; Shenouda, N.; Kongshavn, P.A.; Osmond, D.G. Mechanism of altered B-cell response induced by changes in dietary protein type in mice. J. Nutr. 1985, 115, 1409–1417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Cross, M.L.; Gill, H.S. Modulation of immune function by a modified bovine whey protein concentrate. Immunol. Cell Biol. 1999, 77, 345–350. [Google Scholar] [CrossRef] [PubMed]
  86. Jewell, J.L.; Kim, Y.C.; Russell, R.C.; Yu, F.X.; Park, H.W.; Plouffe, S.W.; Tagliabracci, V.S.; Guan, K.L. Metabolism. Differential regulation of mTORC1 by leucine and glutamine. Science 2015, 347, 194–198. [Google Scholar] [CrossRef] [Green Version]
  87. Moro, T.; Brightwell, C.R.; Velarde, B.; Fry, C.S.; Nakayama, K.; Sanbongi, C.; Volpi, E.; Rasmussen, B.B. Whey protein hydrolysate increases amino acid uptake, mTORC1 signaling, and protein synthesis in skeletal muscle of healthy young men in a randomized crossover trial. J. Nutr. 2019, 149, 1149–1158. [Google Scholar] [CrossRef]
  88. Shimobayashi, M.; Hall, M.N. Multiple amino acid sensing inputs to mTORC1. Cell Res. 2016, 26, 7–20. [Google Scholar] [CrossRef] [Green Version]
  89. Condon, K.J.; Sabatini, D.M. Nutrient regulation of mTORC1 at a glance. J. Cell Sci. 2019, 132, jcs222570. [Google Scholar] [CrossRef]
  90. Melick, C.H.; Jewell. J.L. Regulation of mTORC1 by upstream stimuli. Genes 2020, 11, 989. [Google Scholar] [CrossRef]
  91. Raybuck, A.L.; Lee, K.; Cho, S.H.; Li, J.; Thomas, J.W.; Boothby, M.R. mTORC1 as a cell-intrinsic rheostat that shapes development, preimmune repertoire, and function of B lymphocytes. FASEB J. 2019, 33, 13202–13215. [Google Scholar] [CrossRef] [Green Version]
  92. Davis, T.A.; Nguyen, H.V.; Garcia-Bravo, R.; Fiorotto, M.L.; Jackson, E.M.; Lewis, D.S.; Lee, D.R.; Reeds, P.J. Amino acid composition of human milk is not unique. J. Nutr. 1994, 124, 1126–1132. [Google Scholar] [CrossRef]
  93. Millward, D.J.; Layman, D.K.; Tomé, D.; Schaafsma, G. Protein quality assessment: Impact of expanding understanding of protein and amino acid needs for optimal health. Am. J. Clin. Nutr. 2008, 87, 1576S–1581S. [Google Scholar]
  94. Boirie, Y.; Dangin, M.; Gachon, P.; Vasson, M.P.; Maubois, J.L.; Beaufrère, B. Slow and fast dietary proteins differently modulate postprandial protein accretion. Proc. Natl. Acad. Sci. USA 1997, 94, 14930–14935. [Google Scholar] [CrossRef] [Green Version]
  95. He, T.; Giuseppin, M.L. Slow and fast dietary proteins differentially modulate postprandial metabolism. Int. J. Food Sci. Nutr. 2014, 65, 386–390. [Google Scholar] [CrossRef]
  96. Boutrou, R.; Gaudichon, C.; Dupont, D.; Jardin, J.; Airinei, G.; Marsset-Baglieri, A.; Benamouzig, R.; Tomé, D.; Leonil, J. Sequential release of milk protein-derived bioactive peptides in the jejunum in healthy humans. Am. J. Clin. Nutr. 2013, 97, 1314–1323. [Google Scholar] [CrossRef] [Green Version]
  97. Mahé, S.; Roos, N.; Benamouzig, R.; Davin, L.; Luengo, C.; Gagnon, L.; Gaussergès, N.; Rautureau, J.; Tomé, D. Gastrojejunal kinetics and the digestion of [15N]beta-lactoglobulin and casein in humans: The influence of the nature and quantity of the protein. Am. J. Clin. Nutr. 1996, 63, 546–552. [Google Scholar] [CrossRef] [Green Version]
  98. Kanai, Y.; Segawa, H.; Miyamoto, K.; Uchino, H.; Takeda, E.; Endou, H. Expression cloning and characterization of a transporter for large neutral amino acids activated by the heavy chain of 4F2 antigen (CD98). J. Biol. Chem. 1998, 273, 23629–23632. [Google Scholar] [CrossRef] [Green Version]
  99. Jigjidkhorloo, N.; Kanekura, K.; Matsubayashi, J.; Akahane, D.; Fujita, K.; Oikawa, K.; Kurata, A.; Takanashi, M.; Endou, H.; Nagao, T.; et al. Expression of L-type amino acid transporter 1 is a poor prognostic factor for non-Hodgkin’s lymphoma. Sci. Rep. 2021, 11, 21638. [Google Scholar] [CrossRef]
  100. Le, A.; Lane, A.N.; Hamaker, M.; Bose, S.; Gouw, A.; Barbi, J.; Tsukamoto, T.; Rojas, C.J.; Slusher, B.S.; Zhang, H.; et al. Glucose-independent glutamine metabolism via TCA cycling for proliferation and survival in B cells. Cell Metab. 2012, 15, 110–121. [Google Scholar]
  101. Li, M.; Chiang, Y.L.; Lyssiotis, C.A.; Teater, M.R.; Hong, J.Y.; Shen, H.; Wang, L.; Hu, J.; Jing, H.; Chen, Z.; et al. Non-oncogene addiction to SIRT3 plays a critical role in lymphomagenesis. Cancer Cell. 2019, 35, 916–931. [Google Scholar] [CrossRef]
  102. Durán, R.V.; Hall, M.N. Glutaminolysis feeds mTORC1. Cell Cycle 2012, 11, 4107–4108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Wei, P.; Bott, A.J.; Cluntun, A.A.; Morgan, J.T.; Cunningham, C.N.; Schell, J.C.; Ouyang, Y.; Ficarro, S.B.; Marto, J.A.; Danial, N.N.; et al. Mitochondrial pyruvate supports lymphoma proliferation by fueling a glutamate pyruvate transaminase 2-dependent glutaminolysis pathway. Sci. Adv. 2022, 8, eabq0117. [Google Scholar] [CrossRef] [PubMed]
  104. Li, M.; Teater, M.R.; Hong, J.Y.; Park, N.R.; Duy, C.; Shen, H.; Wang, L.; Chen, Z.; Cerchietti, L.; Davidson, S.M.; et al. Translational activation of ATF4 through mitochondrial anaplerotic metabolic pathways is required for DLBCL growth and survival. Blood Cancer Discov. 2022, 3, 50–65. [Google Scholar] [CrossRef] [PubMed]
  105. Durán, R.V.; Oppliger, W.; Robitaille, A.M.; Heiserich, L.; Skendaj, R.; Gottlieb, E.; Hall, M.N. Glutaminolysis activates Rag-mTORC1 signaling. Mol. Cell. 2012, 47, 349–358. [Google Scholar] [CrossRef] [Green Version]
  106. Lenders, C.M.; Liu, S.; Wilmore, D.W.; Sampson, L.; Dougherty, L.W.; Spiegelman, D.; Willett, W.C. Evaluation of a novel food composition database that includes glutamine and other amino acids derived from gene sequencing data. Eur. J. Clin. Nutr. 2009, 63, 1433–1439. [Google Scholar] [CrossRef]
  107. Baxter, J.H.; Phillips, R.R.; Dowlati, L.; Johns, P.W. Glutamine in commercial liquid nutritional products. J. Agric. Food Chem. 2004, 52, 4963–4968. [Google Scholar] [CrossRef]
  108. Ricci, J.E.; Chiche, J. Metabolic reprogramming of Non-Hodgkin’s B-cell lymphomas and potential therapeutic strategies. Front. Oncol. 2018, 8, 556. [Google Scholar] [CrossRef]
  109. Mlynarczyk, C.; Fontán, L.; Melnick, A. Germinal center-derived lymphomas: The darkest side of humoral immunity. Immunol. Rev. 2019, 288, 214–239. [Google Scholar] [CrossRef] [Green Version]
  110. Fontan, L.; Goldstein, R.; Casalena, G.; Durant, M.; Teater, M.R.; Wilson, J.; Phillip, J.; Xia, M.; Shah, S.; Us, I.; et al. Identification of MALT1 feedback mechanisms enables rational design of potent antilymphoma regimens for ABC-DLBCL. Blood 2021, 137, 788–800. [Google Scholar] [CrossRef]
  111. Ferch, U.; Kloo, B.; Gewies, A.; Pfänder, V.; Düwel, M.; Peschel, C.; Krappmann, D.; Ruland, J. Inhibition of MALT1 protease activity is selectively toxic for activated B cell-like diffuse large B cell lymphoma cells. J. Exp. Med. 2009, 206, 2313–2320. [Google Scholar] [CrossRef]
  112. Hailfinger, S.; Lenz, G.; Ngo, V.; Posvitz-Fejfar, A.; Rebeaud, F.; Guzzardi, M.; Penas, E.M.; Dierlamm, J.; Chan, W.C.; Staudt, L.M.; et al. Essential role of MALT1 protease activity in activated B cell-like diffuse large B-cell lymphoma. Proc. Natl. Acad. Sci. USA 2009, 106, 19946–19951, Erratum in: Proc. Natl. Acad. Sci. USA 2013, 110, 2677. [Google Scholar] [CrossRef] [Green Version]
  113. Phelan, J.D.; Young, R.M.; Webster, D.E.; Roulland, S.; Wright, G.W.; Kasbekar, M.; Shaffer, A.L.; Ceribelli, M.; Wang, J.Q.; Schmitz, R.; et al. A multiprotein supercomplex controlling oncogenic signalling in lymphoma. Nature 2018, 560, 387–391. [Google Scholar] [CrossRef]
  114. Horvilleur, E.; Sbarrato, T.; Hill, K.; Spriggs, R.V.; Screen, M.; Goodrem, P.J.; Sawicka, K.; Chaplin, L.C.; Touriol, C.; Packham, G.; et al. A role for eukaryotic initiation factor 4B overexpression in the pathogenesis of diffuse large B-cell lymphoma. Leukemia 2014, 28, 1092–1102. [Google Scholar] [CrossRef] [Green Version]
  115. Ruggero, D.; Montanaro, L.; Ma, L.; Xu, W.; Londei, P.; Cordon-Cardo, C.; Pandolfi, P.P. The translation factor eIF-4E promotes tumor formation and cooperates with c-Myc in lymphomagenesis. Nat. Med. 2004, 10, 484–486. [Google Scholar] [CrossRef]
  116. Wanner, K.; Hipp, S.; Oelsner, M.; Ringshausen, I.; Bogner, C.; Peschel, C.; Decker, T. Mammalian target of rapamycin inhibition induces cell cycle arrest in diffuse large B cell lymphoma (DLBCL) cells and sensitises DLBCL cells to rituximab. Br. J. Haematol. 2006, 134, 475–484. [Google Scholar] [CrossRef]
  117. Zhao, X.F.; Gartenhaus, R.B. Phospho-p70S6K and cdc2/cdk1 as therapeutic targets for diffuse large B-cell lymphoma. Expert Opin. Ther. Targets 2009, 13, 1085–1093. [Google Scholar] [CrossRef]
  118. Xu, Z.Z.; Xia, Z.G.; Wang, A.H.; Wang, W.F.; Liu, Z.Y.; Chen, L.Y.; Li, J.M. Activation of the PI3K/AKT/mTOR pathway in diffuse large B cell lymphoma: Clinical significance and inhibitory effect of rituximab. Ann. Hematol. 2013, 92, 1351–1358. [Google Scholar] [CrossRef]
  119. Xu, Z.Z.; Wang, W.F.; Fu, W.B.; Wang, A.H.; Liu, Z.Y.; Chen, L.Y.; Guo, P.; Li, J.M. Combination of rituximab and mammalian target of rapamycin inhibitor everolimus (RAD001) in diffuse large B-cell lymphoma. Leuk. Lymphoma 2014, 55, 1151–1157. [Google Scholar] [CrossRef]
  120. Majchrzak, A.; Witkowska, M.; Smolewski, P. Inhibition of the PI3K/Akt/mTOR signaling pathway in diffuse large B-cell lymphoma: Current knowledge and clinical significance. Molecules 2014, 19, 14304–14315. [Google Scholar] [CrossRef] [Green Version]
  121. Merli, M.; Ferrario, A.; Maffioli, M.; Arcaini, L.; Passamonti, F. Everolimus in diffuse large B-cell lymphomas. Future Oncol. 2015, 11, 373–383. [Google Scholar] [CrossRef]
  122. Mao, Y.; Xu, L.; Wang, J.; Zhang, L.; Hou, N.; Xu, J.; Wang, L.; Yang, S.; Chen, Y.; Xiong, L.; et al. ROR1 associates unfavorable prognosis and promotes lymphoma growth in DLBCL by affecting PI3K/Akt/mTOR signaling pathway. Biofactors 2019, 45, 416–426. [Google Scholar] [PubMed]
  123. Taylor, J.; Yeomans, A.M.; Packha, G. Targeted inhibition of mRNA translation initiation factors as a novel therapeutic strategy for mature B-cell neoplasms. Explor. Target Antitumor Ther. 2020, 1, 3–25. [Google Scholar] [PubMed] [Green Version]
  124. Hu, K.; Li, B.; Ma, R.; Yi, H.; Xu, Z.; Peng, Y.; Yu, D.; Wu, H.; Cheng, T.; Lu, Y.; et al. Anti-DLBCL efficacy of DCZ0825 in vitro and in vivo: Involvement of the PI3K–AKT–mTOR/JNK pathway. Acta Biochim. Biophys. Sin. 2021, 53, 575–583. [Google Scholar] [CrossRef] [PubMed]
  125. Yamin, H.; Barnea, M.; Genzer, Y.; Chapnik, N.; Froy, O. Long-term commercial cow’s milk consumption and its effects on metabolic parameters associated with obesity in young mice. Mol. Nutr. Food Res. 2014, 58, 1061–1068. [Google Scholar] [CrossRef] [PubMed]
  126. D’Hulst, G.; Masschelein, E.; De Bock, K. Dampened muscle mTORC1 response following ingestion of high-quality plant-based protein and insect protein compared to whey. Nutrients 2021, 13, 1396. [Google Scholar] [CrossRef]
  127. Tanaka, S.; Baba, Y. B cell receptor signaling. Adv. Exp. Med. Biol. 2020, 1254, 23–36. [Google Scholar]
  128. Küppers, R. Mechanisms of B-cell lymphoma pathogenesis. Nat. Rev. Cancer. 2005, 5, 251–262. [Google Scholar] [CrossRef]
  129. Thurner, L.; Hartmann, S.; Neumann, F.; Hoth, M.; Stilgenbauer, S.; Küppers, R.; Preuss, K.D.; Bewarder, M. Role of specific B-cell receptor antigens in lymphomagenesis. Front. Oncol. 2020, 10, 604685. [Google Scholar]
  130. Thurner, L.; Hartmann, S.; Bewarder, M.; Fadle, N.; Regitz, E.; Schormann, C.; Quiroga, N.; Kemele, M.; Klapper, W.; Rosenwald, A.; et al. Identification of the atypically modified autoantigen Ars2 as the target of B-cell receptors from activated B-cell-type diffuse large B-cell lymphoma. Haematologica 2021, 106, 2224–2232. [Google Scholar]
  131. Wilson, W.H.; Young, R.M.; Schmitz, R.; Yang, Y.; Pittaluga, S.; Wright, G.; Lih, C.J.; Williams, P.M.; Shaffer, A.L.; Gerecitano, J.; et al. Targeting B cell receptor signaling with ibrutinib in diffuse large B cell lymphoma. Nat. Med. 2015, 21, 922–926. [Google Scholar] [CrossRef]
  132. Xu, W.; Berning, P.; Lenz, G. Targeting B-cell receptor and PI3K signaling in diffuse large B-cell lymphoma. Blood 2021, 138, 1110–1119. [Google Scholar]
  133. Wal, J.M. Cow‘s milk proteins/allergens. Ann. Allergy Asthma Immunol. 2002, 89, 3–10. [Google Scholar]
  134. Wal, J.M. Bovine milk allergenicity. Ann. Allergy Asthma Immunol. 2004, 93, S2–S11. [Google Scholar]
  135. Caira, S.; Pinto, G.; Picariello, G.; Vitaglione, P.; De Pascale, S.; Scaloni, A.; Addeo, F. In vivo absorptomics: Identification of bovine milk-derived peptides in human plasma after milk intake. Food Chem. 2022, 385, 132663. [Google Scholar]
  136. Mousan, G.; Kamat, D. Cow’s milk protein allergy. Clin. Pediatr. 2016, 55, 1054–1063. [Google Scholar]
  137. Tedner, S.G.; Asarnoj, A.; Thulin, H.; Westman, M.; Konradsen, J.R.; Nilsson, C. Food allergy and hypersensitivity reactions in children and adults—A review. J. Intern. Med. 2022, 291, 283–302. [Google Scholar]
  138. Lam, H.Y.; van Hoffen, E.; Michelsen, A.; Guikers, K.; van der Tas, C.H.; Bruijnzeel-Koomen, C.A.; Knulst, A.C. Cow’s milk allergy in adults is rare but severe: Both casein and whey proteins are involved. Clin. Exp. Allergy 2008, 38, 995–1002. [Google Scholar]
  139. Laugesen, M.; Elliott, R. Ischaemic heart disease, type 1 diabetes, and cow milk A1 beta-casein. N. Z. Med. J. 2003, 116, U295. [Google Scholar]
  140. Neyestani, T.R.; Djalali, M.; Pezeshki, M.; Siassi, F.; Eshraghian, M.R.; Rajab, A.; Keshavarz, A. Serum antibodies to the major proteins found in cow’s milk of Iranian patients with type 1 diabetes mellitus. Diabetes Nutr. Metab. 2004, 17, 76–83. [Google Scholar]
  141. Monetini, L.; Cavallo, M.G.; Manfrini, S.; Stefanini, L.; Picarelli, A.; Di Tola, M.; Petrone, A.; Bianchi, M.; La Presa, M.; Di Giulio, C.; et al. Antibodies to bovine beta-casein in diabetes and other autoimmune diseases. Horm. Metab. Res. 2002, 34, 455–459. [Google Scholar]
  142. Lamb, M.M.; Miller, M.; Seifert, J.A.; Frederiksen, B.; Kroehl, M.; Rewers, M.; Norris, J.M. The effect of childhood cow’s milk intake and HLA-DR genotype on risk of islet autoimmunity and type 1 diabetes: The Diabetes Autoimmunity Study in the Young. Pediatr. Diabetes 2015, 16, 31–38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Vieira Borba, V.; Lerner, A.; Matthias, T.; Shoenfeld, Y. Bovine milk proteins as a trigger for autoimmune diseases: Myth or reality? Int. J. Celiac Dis. 2020, 8, 10–21. [Google Scholar]
  144. Wang, Y.; Liu, X.; Yan, P.; Bi, Y.; Liu, Y.; Zhang, Z.J. Association between type 1 and type 2 diabetes and risk of non-Hodgkin’s lymphoma: A meta-analysis of cohort studies. Diabetes Metab. 2020, 46, 8–19. [Google Scholar] [PubMed]
  145. Lidén, M.; Kristjánsson, G.; Valtysdottir, S.; Venge, P.; Hällgren, R. Cow’s milk protein sensitivity assessed by the mucosal patch technique is related to irritable bowel syndrome in patients with primary Sjögren’s syndrome. Clin. Exp. Allergy 2008, 38, 929–935. [Google Scholar] [PubMed] [Green Version]
  146. Smedby, K.; Vajdic, C.M.; Falster, M.; Engels, E.A.; Martínez-Maza, O.; Turner, J.; Hjalgrim, H.; Vineis, P.; Seniori Costantini, A.; Bracci, P.M.; et al. Autoimmune disorders and risk of non-Hodgkin lymphoma subtypes: A pooled analysis within the InterLymph Consortium. Blood 2008, 111, 4029–4038. [Google Scholar]
  147. Järvinen, K.M.; Beyer, K.; Vila, L.; Chatchatee, P.; Busse, P.J.; Sampson, H.A. B-cell epitopes as a screening instrument for persistent cow’s milk allergy. J. Allergy Clin. Immunol. 2002, 110, 293–297. [Google Scholar]
  148. Chatchatee, P.; Järvinen, K.M.; Bardina, L.; Vila, L.; Beyer, K.; Sampson, H.A. Identification of IgE and IgG binding epitopes on beta- and kappa-casein in cow’s milk allergic patients. Clin. Exp. Allergy 2001, 31, 1256–1262. [Google Scholar] [CrossRef]
  149. Enomoto, A.; Shon, D.H.; Aoki, Y.; Yamauchi, K.; Kaminogawa, S. Antibodies raised against peptide fragments of bovine alpha s1-casein cross-react with the intact protein only when the peptides contain both B and T cell determinants. Mol. Immunol. 1990, 27, 581–586. [Google Scholar]
  150. Shon, D.H.; Enomoto, A.; Yamauchi, K.; Kaminogawa, S. Antibodies raised against peptide fragments of bovine alpha s1-casein cross-react with the native protein, but recognize sites distinct from the determinants on the protein. Eur. J. Immunol. 1991, 21, 1475–1480. [Google Scholar] [CrossRef]
  151. Fuc, E.; Złotkowska, D.; Stachurska, E.; Wróblewska, B. Immunoreactive properties of α-casein and κ-casein: Ex vivo and in vivo studies. J. Dairy Sci. 2018, 101, 10703–10713. [Google Scholar]
  152. Barati, M.; Jabbari, M.; Fathollahi, M.; Fathollahi, A.; Khaki, V.; Javanmardi, F.; Jazayeri, S.M.H.M.; Shabani, M.; Davoodi, S.H.; Huseyn, E.; et al. Evaluation of different types of milk proteins-derived epitopes using in-silico tools: A primarily study to propose a new definition for bioactive peptides. Food Sci. Technol. Camp. 2022, 42, e102821. [Google Scholar] [CrossRef]
  153. Lozano-Ojalvo, D.; Larré, C.; Klueber, J.; Gelencser, E.; Bueno-Diaz, C.; Diaz-Perales, A.; Benedé, S.; Bavaro, S.L.; Kuehn, A.; Hoffmann-Sommergruber, K.; et al. Are physicochemical properties shaping the allergenic potency of animal allergens? Clin. Rev. Allergy Immunol. 2022, 62, 1–36. [Google Scholar]
  154. Bogahawaththa, D.; Ashraf, R.; Chandrapala, J.; Donkor, O.; Vasiljevic, T. In vitro immunogenicity of various native and thermally processed bovine milk proteins and their mixtures. J. Dairy Sci. 2018, 101, 8726–8736. [Google Scholar] [CrossRef]
  155. Morisawa, Y.; Kitamura, A.; Ujihara, T.; Zushi, N.; Kuzume, K.; Shimanouchi, Y.; Tamura, S.; Wakiguchi, H.; Saito, H.; Matsumoto, K. Effect of heat treatment and enzymatic digestion on the B cell epitopes of cow’s milk proteins. Clin. Exp. Allergy 2009, 39, 918–925. [Google Scholar] [CrossRef]
  156. Teras, L.R.; Patel, A.V.; Hildebrand, J.S.; Gapstur, S.M. Postmenopausal unopposed estrogen and estrogen plus progestin use and risk of non-Hodgkin lymphoma in the American Cancer Society Cancer Prevention Study-II Cohort. Leuk. Lymphoma 2013, 54, 720–725. [Google Scholar]
  157. Hasni, M.S.; Berglund, M.; Yakimchuk, K.; Guan, J.; Linderoth, J.; Amini, R.M.; Enblad, G.; Okret, S. Estrogen receptor β1 in diffuse large B-cell lymphoma growth and as a prognostic biomarker. Leuk. Lymphoma 2017, 58, 418–427. [Google Scholar] [CrossRef]
  158. Faknuam, S.; Assanasen, T.; Ruangvejvorachai, P.; Hanvivadhanakul, P.; Intragumtornchai, T.; Rojnuckarin, P. Estrogen receptor beta expression and prognosis of diffuse large B cell lymphoma. Hematology 2018, 23, 235–241. [Google Scholar]
  159. Langendonk, M.; de Jong, M.R.W.; Smit, N.; Seiler, J.; Reitsma, B.; Ammatuna, E.; Glaudemans, A.W.J.M.; van den Berg, A.; Huls, G.A.; Visser, L.; et al. Identification of the estrogen receptor beta as a possible new tamoxifen-sensitive target in diffuse large B-cell lymphoma. Blood Cancer J. 2022, 12, 36. [Google Scholar] [CrossRef]
  160. Katzenellenbogen, B.S. Biology and receptor interactions of estriol and estriol derivatives in vitro and in vivo. J. Steroid Biochem. 1984, 20, 1033–1037. [Google Scholar] [CrossRef]
  161. Barkhem, T.; Carlsson, B.; Nilsson, Y.; Enmark, E.; Gustafsson, J.; Nilsson, S. Differential response of estrogen receptor alpha and estrogen receptor beta to partial estrogen agonists/antagonists. Mol. Pharmacol. 1998, 54, 105–112. [Google Scholar] [CrossRef]
  162. Paterni, I.; Granchi, C.; Katzenellenbogen, J.A.; Minutolo, F. Estrogen receptors alpha (ERα) and beta (ERβ): Subtype-selective ligands and clinical potential. Steroids 2014, 90, 13–29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Perkins, M.S.; Louw-du Toit, R.; Africander, D. A comparative characterization of estrogens used in hormone therapy via estrogen receptor (ER)-α and -β. J. Steroid Biochem. Mol. Biol. 2017, 174, 27–39. [Google Scholar] [CrossRef] [PubMed]
  164. Mal, R.; Magner, A.; David, J.; Datta, J.; Vallabhaneni, M.; Kassem, M.; Manouchehri, J.; Willingham, N.; Stover, D.; Vandeusen, J.; et al. Estrogen receptor beta (ERβ): A ligand activated tumor suppressor. Front. Oncol. 2020, 10, 587386. [Google Scholar] [CrossRef] [PubMed]
  165. Tang, H.; Liao, Y.; Chen, G.; Xu, L.; Zhang, C.; Ju, S.; Zhou, S. Estrogen upregulates the IGF-1 signaling pathway in lung cancer through estrogen receptor-β. Med. Oncol. 2012, 29, 2640–2648. [Google Scholar] [CrossRef] [PubMed]
  166. Tang, H.; Liao, Y.; Xu, L.; Zhang, C.; Liu, Z.; Deng, Y.; Jiang, Z.; Fu, S.; Chen, Z.; Zhou, S. Estrogen and insulin-like growth factor 1 synergistically promote the development of lung adenocarcinoma in mice. Int. J. Cancer 2013, 133, 2473–2482. [Google Scholar] [CrossRef]
  167. Heap, R.B.; Hamon, M. Oestrone sulphate in milk as an indicator of a viable conceptus in cows. Br. Vet. J. 1979, 135, 355–363. [Google Scholar] [CrossRef]
  168. Snoj, T.; Zuzek, M.C.; Cebulj-Kadunc, N.; Majdic, G. Short communication: Heat treatment and souring do not affect milk estrone and 17β-estradiol concentrations. J. Dairy Sci. 2018, 101, 61–65. [Google Scholar] [CrossRef]
  169. Maruyama, K.; Oshima, T.; Ohyama, K. Exposure to exogenous estrogen through intake of commercial milk produced from pregnant cows. Pediatr. Int. 2010, 52, 28–33. [Google Scholar] [CrossRef]
  170. Farlow, D.W.; Xu, X.; Veenstra, T.D. Comparison of estrone and 17β-estradiol levels in commercial goat and cow milk. J. Dairy Sci. 2012, 95, 1699–1708, Erratum in: J. Dairy Sci. 2012, 95, 4732. [Google Scholar] [CrossRef] [Green Version]
  171. Goyon, A.; Cai, J.Z.; Kraehenbuehl, K.; Hartmann, C.; Shao, B.; Mottier, P. Determination of steroid hormones in bovine milk by LC-MS/MS and their levels in Swiss Holstein cow milk. Food Addit. Contam. Part A Chem. Anal. Control Expo. Risk Assess. 2016, 33, 804–816. [Google Scholar] [CrossRef]
  172. Pape-Zambito, D.A.; Roberts, R.F.; Kensinger, R.S. Estrone and 17beta-estradiol concentrations in pasteurized-homogenized milk and commercial dairy products. J. Dairy Sci. 2010, 93, 2533–2540. [Google Scholar] [CrossRef] [Green Version]
  173. Pape-Zambito, D.A.; Magliaro, A.L.; Kensinger, R.S. Concentrations of 17beta-estradiol in Holstein whole milk. J. Dairy Sci. 2007, 90, 3308–3313. [Google Scholar] [CrossRef] [Green Version]
  174. Hartmann, S.; Lacorn, M.; Steinhart, H. Natural occurrence of steroid hormones in food. Food Chem. 1989, 62, 7–20. [Google Scholar] [CrossRef]
  175. Cavaliere, C.; Capriotti, A.L.; Foglia, P.; Piovesana, S.; Samperi, R.; Ventura, S.; Laganà, A. Natural estrogens in dairy products: Determination of free and conjugated forms by ultra high performance liquid chromatography with tandem mass spectrometry. J. Sep. Sci. 2015, 38, 3599–3606. [Google Scholar] [CrossRef]
  176. Fenichel, P.; Chevalier, N.; Brucker-Davis, F. Bisphenol A: An endocrine and metabolic disruptor. Ann. Endocrinol. 2013, 74, 211–220. [Google Scholar] [CrossRef]
  177. Rochester, J.R. Bisphenol A and human health: A review of the literature. Reprod. Toxicol. 2013, 42, 132–155. [Google Scholar] [CrossRef]
  178. Abraham, A.; Chakraborty, P. A review on sources and health impacts of bisphenol A. Rev. Environ. Health 2020, 35, 201–210. [Google Scholar] [CrossRef]
  179. Rogers, J.A.; Metz, L.; Yong, V.W. Review: Endocrine disrupting chemicals and immune responses: A focus on bisphenol-A and its potential mechanisms. Mol. Immunol. 2013, 53, 421–430. [Google Scholar]
  180. Kimber, I. Bisphenol A and immunotoxic potential: A commentary. Regul. Toxicol. Pharmacol. 2017, 90, 358–363. [Google Scholar] [CrossRef]
  181. Segovia-Mendoza, M.; Nava-Castro, K.E.; Palacios-Arreola, M.I.; Garay-Canales, C.; Morales-Montor, J. How microplastic components influence the immune system and impact on children health: Focus on cancer. Birth Defects Res. 2020, 12, 1341–1361. [Google Scholar] [CrossRef]
  182. Ge, X.; Lv, X.; Feng, L.; Liu, X.; Wang, X. High expression and nuclear localization of β-catenin in diffuse large B-cell lymphoma. Mol. Med. Rep. 2012, 5, 1433–1437. [Google Scholar] [PubMed] [Green Version]
  183. Zhang, D.Y.; Wang, H.J.; Tan, Y.Z. Wnt/β-catenin signaling induces the aging of mesenchymal stem cells through the DNA damage response and the p53/p21 pathway. PLoS ONE 2011, 6, e21397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Derenzini, E.; Agostinelli, C.; Imbrogno, E.; Iacobucci, I.; Casadei, B.; Brighenti, E.; Righi, S.; Fuligni, F.; Ghelli Luserna Di Rorà, A.; Ferrari, A.; et al. Constitutive activation of the DNA damage response pathway as a novel therapeutic target in diffuse large B-cell lymphoma. Oncotarget 2015, 6, 6553–6569. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Dupont, J.; Fernandez, A.M.; Glackin, C.A.; Helman, L.; LeRoith, D. Insulin-like growth factor 1 (IGF-1)-induced twist expression is involved in the anti-apoptotic effects of the IGF-1 receptor. J. Biol. Chem. 2001, 276, 26699–26707. [Google Scholar] [CrossRef] [Green Version]
  186. Lemma, S.; Karihtala, P.; Haapasaari, K.M.; Jantunen, E.; Soini, Y.; Bloigu, R.; Pasanen, A.K.; Turpeenniemi-Hujanen, T.; Kuittinen, O. Biological roles and prognostic values of the epithelial-mesenchymal transition-mediating transcription factors Twist, ZEB1 and Slug in diffuse large B-cell lymphoma. Histopathology 2013, 62, 326–333. [Google Scholar] [CrossRef]
  187. Davis, R.E.; Brown, K.D.; Siebenlist, U.; Staudt, L.M. Constitutive nuclear factor kappaB activity is required for survival of activated B cell-like diffuse large B cell lymphoma cells. J. Exp. Med. 2001, 194, 1861–1874. [Google Scholar] [CrossRef]
  188. Compagno, M.; Lim, W.K.; Grunn, A.; Nandula, S.V.; Brahmachary, M.; Shen, Q.; Bertoni, F.; Ponzoni, M.; Scandurra, M.; Califano, A.; et al. Mutations of multiple genes cause deregulation of NF-kappaB in diffuse large B-cell lymphoma. Nature 2009, 459, 717–721. [Google Scholar] [CrossRef] [Green Version]
  189. Nagel, D.; Vincendeau, M.; Eitelhuber, A.C.; Krappmann, D. Mechanisms and consequences of constitutive NF-κB activation in B-cell lymphoid malignancies. Oncogene 2014, 33, 5655–5665. [Google Scholar] [CrossRef] [Green Version]
  190. Zhu, J.; Jiang, L.; Liu, Y.; Qian, W.; Liu, J.; Zhou, J.; Gao, R.; Xiao, H.; Wang, J. MAPK and NF-κB pathways are involved in bisphenol A-induced TNF-α and IL-6 production in BV2 microglial cells. Inflammation 2015, 38, 637–648. [Google Scholar] [CrossRef]
  191. Jalal, N.; Surendranath, A.R.; Pathak, J.L.; Yu, S.; Chung, C.Y. Bisphenol A (BPA) the mighty and the mutagenic. Toxicol. Rep. 2017, 5, 76–84. [Google Scholar] [CrossRef]
  192. Yu, M.; Xu, Y.; Li, M.; Li, D.; Lu, Y.; Yu, D.; Du, W. Bisphenol A accelerates meiotic progression in embryonic chickens via the estrogen receptor β signaling pathway. Gen. Comp. Endocrinol. 2018, 259, 66–75. [Google Scholar] [CrossRef]
  193. Burkhardt, F.; Schulz, S.D.; Hellwig, E.; Vach, K.; Tomakidi, P.; Polydorou, O. Low-dose bisphenol A and its analogues bisphenol F and S activate estrogen receptor ß and slightly modulate genes in human gingival keratinocytes. Dent. Mater. 2021, 37, 625–635. [Google Scholar] [CrossRef]
  194. Wen, X.; Zhu, M.; Li, Z.; Li, T.; Xu, X. Dual effects of bisphenol A on wound healing, involvement of estrogen receptor β. Ecotoxicol. Environ. Saf. 2022, 231, 113207. [Google Scholar] [CrossRef]
  195. Zhang, Y.; Wei, F.; Zhang, J.; Hao, L.; Jiang, J.; Dang, L.; Mei, D.; Fan, S.; Yu, Y.; Jiang, L. Bisphenol A and estrogen induce proliferation of human thyroid tumor cells via an estrogen-receptor-dependent pathway. Arch. Biochem. Biophys. 2017, 633, 29–39. [Google Scholar] [CrossRef]
  196. Farahani, M.; Rezaei-Tavirani, M.; Arjmand, B. A systematic review of microRNA expression studies with exposure to bisphenol A. J. Appl. Toxicol. 2021, 41, 4–19. [Google Scholar] [CrossRef]
  197. Ferrante, M.; Cristaldi, A.; Oliveri Conti, G. Oncogenic role of miRNA in environmental exposure to plasticizers: A systematic review. J. Pers. Med. 2021, 11, 500. [Google Scholar] [CrossRef]
  198. Hui, L.; Li, H.; Lu, G.; Chen, Z.; Sun, W.; Shi, Y.; Fu, Z.; Huang, B.; Zhu, X.; Lu, W.; et al. Low dose of bisphenol A modulates ovarian cancer gene expression profile and promotes epithelial to mesenchymal transition via canonical Wnt pathway. Toxicol. Sci. 2018, 164, 527–538. [Google Scholar] [CrossRef]
  199. Sabry, R.; Saleh, A.C.; Stalker, L.; LaMarre, J.; Favetta, L.A. Effects of bisphenol A and bisphenol S on microRNA expression during bovine (Bos taurus) oocyte maturation and early embryo development. Reprod. Toxicol. 2021, 99, 96–108. [Google Scholar] [CrossRef]
  200. Oldenburg, J.; Fürhacker, M.; Hartmann, C.; Steinbichl, P.; Banaderakhshan, R.; Haslberger, A. Different bisphenols induce non-monotonous changes in miRNA expression and LINE-1 methylation in two cell lines. Environ. Epigenet. 2021, 7, dvab011. [Google Scholar] [CrossRef]
  201. Mercogliano, R.; Santonicola, S. Investigation on bisphenol A levels in human milk and dairy supply chain: A review. Food Chem. Toxicol. 2018, 114, 98–107. [Google Scholar] [CrossRef]
  202. Herrero, L.; Quintanilla-López, J.E.; Fernández, M.A.; Gómara, B. Plasticisers and preservatives in commercial milk products: A comprehensive study on packages used in the Spanish market. Food Chem. 2021, 338, 128031. [Google Scholar] [PubMed]
  203. Santonicola, S.; Ferrante, M.C.; Murru, N.; Gallo, P.; Mercogliano, R. Hot topic: Bisphenol A in cow milk and dietary exposure at the farm level. J. Dairy Sci. 2019, 102, 1007–1013. [Google Scholar] [PubMed] [Green Version]
  204. Mercogliano, R.; Santonicola, S.; Albrizio, S.; Ferrante, M.C. Occurrence of bisphenol A in the milk chain: A monitoring model for risk assessment at a dairy company. J. Dairy Sci. 2021, 104, 5125–5132. [Google Scholar] [PubMed]
  205. Farooq, M.U.; Jalees, M.I.; Qurat-Ul-Ain, H.G.; Anis, M.; Islam, U. Health risk assessment of endocrine disruptor bisphenol A leaching from plastic bottles of milk and soft drinks. Environ. Sci. Pollut. Res. Int. 2021, 28, 57090–57098. [Google Scholar] [PubMed]
  206. Högfeldt, T.; Jaing, C.; Loughlin, K.M.; Thissen, J.; Gardner, S.; Bahnassy, A.A.; Gharizadeh, B.; Lundahl, J.; Österborg, A.; Porwit, A.; et al. Differential expression of viral agents in lymphoma tissues of patients with ABC diffuse large B-cell lymphoma from high and low endemic infectious disease regions. Oncol. Lett. 2016, 12, 2782–2788. [Google Scholar] [CrossRef] [Green Version]
  207. Bilajac, E.; Mahmutović, L.; Lundstrom, K.; Glamočlija, U.; Šutković, J.; Sezer, A.; Hromić-Jahjefendić, A. Viral agents as potential drivers of diffuse large B-Cell lymphoma tumorigenesis. Viruses 2022, 14, 2105. [Google Scholar]
  208. Hong, J.Y.; Ryu, K.J.; Park, C.; Hong, M.; Ko, Y.H.; Kim, W.S.; Kim, S.J. Clinical impact of serum survivin positivity and tissue expression of EBV-encoded RNA in diffuse large B-cell lymphoma patients treated with rituximab-CHOP. Oncotarget 2017, 8, 13782–13791. [Google Scholar]
  209. Castillo, J.J.; Beltran, B.E.; Miranda, R.N.; Young, K.H.; Chavez, J.C.; Sotomayor, E.M. EBV-positive diffuse large B-cell lymphoma, not otherwise specified: 2018 update on diagnosis, risk-stratification and management. Am. J. Hematol. 2018, 93, 953–962. [Google Scholar]
  210. Beltran, B.E.; Castro, D.; Paredes, S.; Miranda, R.N.; Castillo, J.J. EBV-positive diffuse large B-cell lymphoma, not otherwise specified: 2020 update on diagnosis, risk-stratification and management. Am. J. Hematol. 2020, 95, 435–445. [Google Scholar]
  211. Marques-Piubelli, M.L.; Salas, Y.I.; Pachas, C.; Becker-Hecker, R.; Vega, F.; Miranda, R.N. Epstein-Barr virus-associated B-cell lymphoproliferative disorders and lymphomas: A review. Pathology 2020, 52, 40–52. [Google Scholar]
  212. Shibusawa, M.; Kidoguchi, K.; Tanimoto, T. Chapter 2: Epstein-Barr Virus-Positive Diffuse Large B Cell Lymphoma. In Lymphoma; Gallamini, A., Juweid, M., Eds.; Exon Publications: Brisbane, Australia, 2021. [Google Scholar]
  213. Zhao, C.X.; Wen, J.J.; Fu, D.; Xu, P.P.; Cheng, S.; Wang, L.; Wang, C.F.; Fei, X.C.; Wang, X.; Zhou, J.F.; et al. Clinical and molecular features of Epstein-Barr virus-positive diffuse large B-cell lymphoma: Results in a multi-center trial. Clin. Transl. Med. 2021, 11, e539. [Google Scholar] [CrossRef]
  214. Chabay, P. Advances in the pathogenesis of EBV-associated diffuse large B cell lymphoma. Cancers 2021, 13, 2717. [Google Scholar]
  215. Satou, A.; Nakamura, S. EBV-positive B-cell lymphomas and lymphoproliferative disorders: Review from the perspective of immune escape and immunodeficiency. Cancer Med. 2021, 10, 6777–6785. [Google Scholar]
  216. Malpica, L.; Marques-Piubelli, M.L.; Beltran, B.E.; Chavez, J.C.; Miranda, R.N.; Castillo, J.J. EBV-positive diffuse large B-cell lymphoma, not otherwise specified: 2022 update on diagnosis, risk-stratification, and management. Am. J. Hematol. 2022, 97, 951–965. [Google Scholar]
  217. Carbone, A.; Cesarman, E.; Spina, M.; Gloghini, A.; Schulz, T.F. HIV-associated lymphomas and gamma-herpesviruses. Blood 2009, 113, 1213–1224. [Google Scholar]
  218. Gloghini, A.; Dolcetti, R.; Carbone, A. Lymphomas occurring specifically in HIV-infected patients: From pathogenesis to pathology. Semin. Cancer Biol. 2013, 23, 457–467. [Google Scholar]
  219. Baptista, M.J.; Garcia, O.; Morgades, M.; Gonzalez-Barca, E.; Miralles, P.; Lopez-Guillermo, A.; Abella, E.; Moreno, M.; Sancho, J.M.; Feliu, E.; et al. HIV-infection impact on clinical-biological features and outcome of diffuse large B-cell lymphoma treated with R-CHOP in the combination antiretroviral therapy era. AIDS 2015, 29, 811–818. [Google Scholar]
  220. Besson, C.; Lancar, R.; Prevot, S.; Algarte-Genin, M.; Delobel, P.; Bonnet, F.; Meyohas, M.C.; Partisani, M.; Oberic, L.; Gabarre, J.; et al. Outcomes for HIV-associated diffuse large B-cell lymphoma in the modern combined antiretroviral therapy era. AIDS 2017, 31, 2493–2501. [Google Scholar]
  221. Wu, J.; Miao, Y.; Qian, C.; Tao, P.; Wang, X.; Dong, X.; Li, X.; Lou, J.; Liang, J.; Xu, W.; et al. Clinical characteristics and outcomes in HIV-associated diffuse large B-cell lymphoma in China: A retrospective single-center study. J. Cancer 2021, 12, 2903–2911. [Google Scholar] [CrossRef]
  222. Ren, W.; Ye, X.; Su, H.; Li, W.; Liu, D.; Pirmoradian, M.; Wang, X.; Zhang, B.; Zhang, Q.; Chen, L.; et al. Genetic landscape of hepatitis B virus-associated diffuse large B-cell lymphoma. Blood 2018, 131, 2670–2681, Erratum: Blood 2019, 133, 620. [Google Scholar] [CrossRef]
  223. Wang, Y.; Wang, H.; Pan, S.; Hu, T.; Shen, J.; Zheng, H.; Xie, S.; Xie, Y.; Lu, R.; Guo, L. Capable infection of hepatitis B virus in diffuse large B-cell lymphoma. J. Cancer 2018, 9, 1575–1581. [Google Scholar] [CrossRef] [PubMed]
  224. Visentini, M.; Casato, M. HBV messing with the B-cell genome leads to DLBCL. Blood 2018, 131, 2602–2603. [Google Scholar] [CrossRef] [PubMed]
  225. Zhou, X.; Wuchter, P.; Egerer, G.; Kriegsmann, M.; Kommoss, F.K.F.; Witzens-Harig, M.; Kriegsmann, K. Serological hepatitis B virus (HBV) activity in patients with HBV infection and B-cell non-Hodgkin’s lymphoma. Eur. J. Haematol. 2020, 104, 469–475. [Google Scholar] [CrossRef] [PubMed]
  226. Ye, X.; Wang, L.; Nie, M.; Wang, Y.; Dong, S.; Ren, W.; Li, G.; Li, Z.M.; Wu, K.; Pan-Hammarström, Q. A single-cell atlas of diffuse large B cell lymphoma. Cell Rep. 2022, 39, 110713. [Google Scholar] [CrossRef] [PubMed]
  227. Visco, C.; Finotto, S. Hepatitis C virus and diffuse large B-cell lymphoma: Pathogenesis, behavior and treatment. World J. Gastroenterol. 2014, 20, 11054–11061. [Google Scholar] [CrossRef]
  228. Saleh, L.M.; Canioni, D.; Shamaa, S.; El-Zaafarany, M.; Emarah, Z.; Abdel-Aziz, S.; Eladle, E.; Abdelaziz, A.; Hermine, O.; Besson, C.; et al. High prevalence of hepatitis C virus among B-cell non Hodgkin lymphoma patients in Mansoura Region (Egypt), ANRS 12263 Study. Mediterr. J. Hematol. Infect. Dis. 2019, 11, e2019011. [Google Scholar] [CrossRef] [Green Version]
  229. Merli, M.; Frigeni, M.; Alric, L.; Visco, C.; Besson, C.; Mannelli, L.; Di Rocco, A.; Ferrari, A.; Farina, L.; Pirisi, M.; et al. Direct-acting antivirals in hepatitis C virus-associated diffuse large B-cell lymphomas. Oncologist 2019, 24, e720–e729. [Google Scholar] [CrossRef] [Green Version]
  230. Beltran, B.E.; Quiñones, P.; Morales, D.; Revilla, J.C.; Alva, J.C.; Castillo, J.J. Diffuse large B-cell lymphoma in human T-lymphotropic virus type 1 carriers. Leuk. Res. Treat. 2012, 2012, 262363. [Google Scholar] [CrossRef]
  231. Amara, K.; Trimeche, M.; Ziadi, S.; Laatiri, A.; Hachana, M.; Sriha, B.; Mokni, M.; Korbi, S. Presence of simian virus 40 DNA sequences in diffuse large B-cell lymphomas in Tunisia correlates with aberrant promoter hypermethylation of multiple tumor suppressor genes. Int. J. Cancer 2007, 121, 2693–2702. [Google Scholar] [CrossRef]
  232. Amara, K.; Trimeche, M.; Ziadi, S.; Laatiri, A.; Mestiri, S.; Sriha, B.; Mokni, M.; Korbi, S. Presence of simian virus 40 in diffuse large B-cell lymphomas in Tunisia correlates with germinal center B-cell immunophenotype, t(14;18) translocation, and P53 accumulation. Mod. Pathol. 2008, 21, 282–296. [Google Scholar] [CrossRef] [Green Version]
  233. Samaka, R.M.; Aiad, H.A.; Kandil, M.A.; Asaad, N.Y.; Holah, N.S. The prognostic role and relationship between E2F1 and SV40 in diffuse large B-cell lymphoma of Egyptian patients. Anal. Cell Pathol. 2015, 2015, 919834. [Google Scholar] [CrossRef] [Green Version]
  234. Calabrò, M.L.; Sarid, R. Human herpesvirus 8 and lymphoproliferative disorders. Mediterr. J. Hematol. Infect. Dis. 2018, 10, e2018061. [Google Scholar] [CrossRef]
  235. Wen, K.W.; Wang, L.; Menke, J.R.; Damania, B. Cancers associated with human gammaherpesviruses. FEBS J. 2022, 289, 7631–7669. [Google Scholar] [CrossRef]
  236. Fenu, E.M.; Beaty, M.W.; O’Neill, T.E.; O’Neill, S.S. Cardiac involvement by human herpesvirus 8-positive diffuse large B-cell lymphoma: An unusual presentation in a patient with human immunodeficiency virus. Case Rep. Pathol. 2022, 2022, 1298121. [Google Scholar] [CrossRef]
  237. Sreehari, S. High risk HPV, HSIL and primary diffuse large B cell lymphoma of cervix: An unsual case. IJCSMB 2019, 5, 555666. [Google Scholar] [CrossRef]
  238. Ren, X.; Cheng, Y.; Wu, S.; Zeng, X.; Shi, X.; Ling, Q.; Li, Z.; Liang, Z.; Wang, B. Primary non-Hodgkin lymphoma of the tongue base: The clinicopathology of seven cases and evaluation of HPV and EBV status. Diagn. Pathol. 2020, 15, 30. [Google Scholar] [CrossRef] [Green Version]
  239. Swerdlow, S.H.; Campo, E.; Pileri, S.A.; Harris, N.L.; Stein, H.; Siebert, R.; Advani, R.; Ghielmini, M.; Salles, G.A.; Zelenetz, A.D.; et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood 2016, 127, 2375–2390. [Google Scholar] [CrossRef] [Green Version]
  240. Linnstaedt, S.D.; Gottwein, E.; Skalsky, R.L.; Luftig, M.A.; Culle, B.R. Virally induced cellular microRNA miR-155 plays a key role in B-cell immortalization by Epstein-Barr virus. J. Virol. 2010, 84, 11670–11678. [Google Scholar] [CrossRef] [Green Version]
  241. Wood, C.D.; Carvell, T.; Gunnell, A.; Ojeniyi, O.O.; Osborne, C.; West, M.J. Enhancer control of microRNA miR-155 expression in Epstein-Barr virus-infected B cells. J. Virol. 2018, 92, e00716–e00718, Erratum in: J. Virol. 2019, 93, 3. [Google Scholar] [CrossRef] [Green Version]
  242. Gottwein, E.; Mukherjee, N.; Sachse, C.; Frenzel, C.; Majoros, W.H.; Chi, J.T.; Braich, R.; Manoharan, M.; Soutschek, J.; Ohler, U.; et al. A viral microRNA functions as an orthologue of cellular miR-155. Nature 2007, 450, 1096–1099. [Google Scholar] [CrossRef] [Green Version]
  243. Sekar, D.; Hairul Islam, V.I.; Thirugnanasambantham, K.; Saravanan, S. Relevance of miR-21 in HIV and non-HIV-related lymphomas. Tumour Biol. 2014, 35, 8387–8393. [Google Scholar] [CrossRef] [PubMed]
  244. Thapa, D.R.; Bhatia, K.; Bream, J.H.; D’Souza, G.; Rinaldo, C.R.; Wolinsky, S.; Detels, R.; Martínez-Maza, O. B-cell activation induced microRNA-21 is elevated in circulating B cells preceding the diagnosis of AIDS-related non-Hodgkin lymphomas. AIDS 2012, 26, 1177–1180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  245. He, G.; Ding, J.; Zhang, Y.; Cai, M.; Yang, J.; Cho, W.C.; Zheng, Y. microRNA-21: A key modulator in oncogenic viral infections. RNA Biol. 2021, 18, 809–817. [Google Scholar] [CrossRef] [PubMed]
  246. Wu, J.; Yang, J.; Ding, J.; Guo, X.; Zhu, X.Q.; Zheng, Y. Exosomes in virus-associated cancer. Cancer Lett. 2018, 438, 44–51. [Google Scholar] [CrossRef]
  247. Krupovic, M.; Varsani, A.; Kazlauskas, D.; Breitbart, M.; Delwart, E.; Rosario, K.; Yutin, N.; Wolf, Y.I.; Harrach, B.; Zerbini, F.M.; et al. Cressdnaviricota: A virus phylum unifying seven families of Rep-encoding viruses with single-stranded, circular DNA genomes. J. Virol. 2020, 94, e00582-20. [Google Scholar] [CrossRef]
  248. Zur Hausen, H.; de Villiers, E.M. Dairy cattle serum and milk factors contributing to the risk of colon and breast cancers. Int. J. Cancer 2015, 137, 959–967. [Google Scholar] [CrossRef]
  249. Zur Hausen, H.; Bund, T.; de Villiers, E.M. Infectious agents in bovine red meat and milk and their potential role in cancer and other chronic diseases. Curr. Top. Microbiol. Immunol. 2017, 407, 83–116. [Google Scholar]
  250. Zur Hausen, H.; Bund, T.; de Villiers, E.M. Specific nutritional infections early in life as risk factors for human colon and breast cancers several decades later. Int. J. Cancer 2019, 144, 1574–1583. [Google Scholar] [CrossRef]
  251. Bund, T.; Nikitina, E.; Chakraborty, D.; Ernst, C.; Gunst, K.; Boneva, B.; Tessmer, C.; Volk, N.; Brobeil, A.; Weber, A.; et al. Analysis of chronic inflammatory lesions of the colon for BMMF Rep antigen expression and CD68 macrophage interactions. Proc. Natl. Acad. Sci. USA 2021, 118, e2025830118. [Google Scholar] [CrossRef]
  252. Nikitina, E.; Alikhanyan, K.; Neßling, M.; Richter, K.; Kaden, S.; Ernst, C.; Seitz, S.; Chuprikova, L.; Häfele, L.; Gunst, K.; et al. Structural expression of bovine milk and meat factors in tissues of colorectal, lung and pancreatic cancer patients. Int. J. Cancer 2022. [Google Scholar] [CrossRef]
  253. de Villiers, E.M.; Gunst, K.; Chakraborty, D.; Ernst, C.; Bund, T.; Zur Hausen, H. A specific class of infectious agents isolated from bovine serum and dairy products and peritumoral colon cancer tissue. Emerg. Microbes Infect. 2019, 8, 1205–1218. [Google Scholar] [CrossRef] [Green Version]
  254. König, M.T.; Fux, R.; Link, E.; Sutter, G.; Märtlbauer, E.; Didier, A. Identification and characterization of circular single-stranded DNA genomes in sheep and goat milk. Viruses 2021, 13, 2176. [Google Scholar] [CrossRef]
  255. König, M.T.; Fux, R.; Link, E.; Sutter, G.; Märtlbauer, E.; Didier, A. Circular Rep-encoding single-stranded DNA sequences in milk from water buffaloes (Bubalus arnee f. bubalis). Viruses 2021, 13, 1088. [Google Scholar] [CrossRef]
  256. de Villiers, E.M.; Zur Hausen, H. Bovine meat and milk factors (BMMFs): Their proposed role in common human cancers and type 2 diabetes mellitus. Cancers 2021, 13, 5407. [Google Scholar] [CrossRef]
  257. Ashrafi, F.; Nassiri, M.; Javadmanesh, A.; Rahimi, H.; Rezaee, S.A. Epigenetics evaluation of the oncogenic mechanisms of two closely related bovine and human deltaretroviruses: A system biology study. Microb. Pathog. 2020, 139, 103845. [Google Scholar] [CrossRef]
  258. Marawan, M.A.; Alouffi, A.; El Tokhy, S.; Badawy, S.; Shirani, I.; Dawood, A.; Guo, A.; Almutairi, M.M.; Alshammari, F.A.; Selim, A. Bovine leukaemia virus: Current epidemiological circumstance and future prospective. Viruses 2021, 13, 2167. [Google Scholar] [CrossRef]
  259. Ferrer, J.F.; Piper, C.E. An evaluation of the role of milk in the natural transmission of BLV. Ann. Rech. Vet. 1978, 9, 803–807. [Google Scholar]
  260. Ferrer, J.F.; Piper, C.E. Role of colostrum and milk in the natural transmission of the bovine leukemia virus. Cancer Res. 1981, 41, 4906–4909. [Google Scholar]
  261. Corredor-Figueroa, A.P.; Olaya-Galán, N.N.; Velandia-Álvarez, S.; Muñoz, M.; Salas-Cárdenas, S.P.; Ibáñez-Pinilla, M.; Patarroyo, M.A.; Gutiérrez, M.F. Co-circulation of bovine leukemia virus haplotypes among humans, animals, and food products: New insights of its zoonotic potential. Int. J. Environ. Res. Public Health 2021, 18, 4883. [Google Scholar] [CrossRef]
  262. Olaya-Galán, N.N.; Corredor-Figueroa, A.P.; Guzmán-Garzón, T.C.; Ríos-Hernandez, K.S.; Salas-Cárdenas, S.P.; Patarroyo, M.A.; Gutierrez, M.F. Bovine leukaemia virus DNA in fresh milk and raw beef for human consumption. Epidemiol. Infect. 2017, 145, 3125–3130. [Google Scholar] [CrossRef] [Green Version]
  263. Buehring, G.C.; DeLaney, A.; Shen, H.; Chu, D.L.; Razavian, N.; Schwartz, D.A.; Demkovich, Z.R.; Bates, M.N. Bovine leukemia virus discovered in human blood. BMC Infect. Dis. 2019, 19, 297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  264. Buehring, G.C.; Shen, H.M.; Jensen, H.M.; Choi, K.Y.; Sun, D.; Nuovo, G. Bovine leukemia virus DNA in human breast tissue. Emerg. Infect. Dis. 2014, 20, 772–782. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  265. Khan, Z.; Abubakar, M.; Arshed, M.J.; Aslam, R.; Sattar, S.; Shah, N.A.; Javed, S.; Tariq, A.; Bostan, N.; Manzoor, S. Molecular investigation of possible relationships concerning bovine leukemia virus and breast cancer. Sci. Rep. 2022, 12, 4161. [Google Scholar] [CrossRef] [PubMed]
  266. Kincaid, R.P.; Burke, J.M.; Sullivan, C.S. RNA virus microRNA that mimics a B-cell oncomiR. Proc. Natl. Acad. Sci. USA 2012, 109, 3077–3082. [Google Scholar] [CrossRef] [Green Version]
  267. Souza, O.F.; Popi, A.F. Role of microRNAs in B-cell compartment: Development, proliferation and hematological diseases. Biomedicine 2022, 10, 2004. [Google Scholar] [CrossRef]
  268. Silva, M.; Melo, S.A. Non-coding RNAs in exosomes: New players in cancer biology. Curr. Genomics 2015, 16, 295–303. [Google Scholar] [CrossRef] [Green Version]
  269. Hanahan, D. Hallmarks of cancer: New dimensions. Cancer Discov. 2022, 12, 31–46. [Google Scholar] [CrossRef]
  270. Coffre, M.; Koralov, S.B. miRNAs in B cell development and lymphomagenesis. Trends Mol. Med. 2017, 23, 721–736. [Google Scholar] [CrossRef]
  271. Fuertes, T.; Salgado, I.; de Yébenes, V.G. microRNA fine-tuning of the germinal center response. Front. Immunol. 2021, 12, 660450. [Google Scholar] [CrossRef]
  272. Shi, Y.; Ding, D.; Qu, R.; Tang, Y.; Hao, S. Non-coding RNAs in diffuse large B-cell lymphoma. Onco Targets Ther. 2020, 13, 12097–12112. [Google Scholar] [CrossRef]
  273. Yazdanparast, S.; Huang, Z.; Keramat, S.; Izadirad, M.; Li, Y.D.; Bo, L.; Gharehbaghian, A.; Chen, Z.S. The roles of exosomal microRNAs in diffuse large B-cell lymphoma: Diagnosis, prognosis, clinical application, and biomolecular mechanisms. Front. Oncol. 2022, 12, 904637. [Google Scholar] [CrossRef]
  274. Rutherford, S.C.; Fachel, A.A.; Li, S.; Sawh, S.; Muley, A.; Ishii, J.; Saxena, A.; Dominguez, P.M.; Caldas Lopes, E.; Agirre, X.; et al. Extracellular vesicles in DLBCL provide abundant clues to aberrant transcriptional programming and genomic alterations. Blood 2018, 132, e13–e23. [Google Scholar] [CrossRef]
  275. Alderton, G.K. Metastasis. Exosomes drive premetastatic niche formation. Nat. Rev. Cancer 2012, 12, 447. [Google Scholar] [CrossRef]
  276. Ghosh, A.K.; Secreto, C.R.; Knox, T.R.; Ding, W.; Mukhopadhyay, D.; Kay, N.E. Circulating microvesicles in B-cell chronic lymphocytic leukemia can stimulate marrow stromal cells: Implications for disease progression. Blood 2010, 115, 1755–1764. [Google Scholar] [CrossRef] [Green Version]
  277. Peinado, H.; Alečković, M.; Lavotshkin, S.; Matei, I.; Costa-Silva, B.; Moreno-Bueno, G.; Hergueta-Redondo, M.; Williams, C.; García-Santos, G.; Ghajar, C.; et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat. Med. 2012, 18, 883–891, Erratum in: Nat. Med. 2016, 22, 1502. [Google Scholar] [CrossRef] [Green Version]
  278. Melnik, B.C.; John, S.M.; Carrera-Bastos, P.; Schmitz, G. MicroRNA-21-enriched exosomes as epigenetic regulators in melanomagenesis and melanoma progression: The impact of Western lifestyle factors. Cancers 2020, 12, 2111. [Google Scholar] [CrossRef]
  279. Costa-Silva, B.; Aiello, N.M.; Ocean, A.J.; Singh, S.; Zhang, H.; Thakur, B.K.; Becker, A.; Hoshino, A.; Mark, M.T.; Molina, H.; et al. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat. Cell Biol. 2015, 17, 816–826. [Google Scholar] [CrossRef]
  280. Lin, Q.; Zhou, C.R.; Bai, M.J.; Zhu, D.; Chen, J.W.; Wang, H.F.; Li, M.A.; Wu, C.; Li, Z.R.; Huang, M.S. Exosome-mediated miRNA delivery promotes liver cancer EMT and metastasis. Am. J. Transl. Res. 2020, 12, 1080–1095. [Google Scholar]
  281. Melnik, B.C. Dairy consumption and hepatocellular carcinoma risk. Ann. Transl. Med. 2021, 9, 736. [Google Scholar] [CrossRef]
  282. Yuan, X.; Qian, N.; Ling, S.; Li, Y.; Sun, W.; Li, J.; Du, R.; Zhong, G.; Liu, C.; Yu, G.; et al. Breast cancer exosomes contribute to pre-metastatic niche formation and promote bone metastasis of tumor cells. Theranostics 2021, 11, 1429–1445. [Google Scholar] [CrossRef]
  283. Chen, X.; Feng, J.; Chen, W.; Shao, S.; Chen, L.; Wan, H. Small extracellular vesicles: From promoting pre-metastatic niche formation to therapeutic strategies in breast cancer. Cell Commun. Signal. 2022, 20, 141. [Google Scholar] [CrossRef] [PubMed]
  284. Saber, S.H.; Ali, H.E.A.; Gaballa, R.; Gaballah, M.; Ali, H.I.; Zerfaoui, M.; Abd Elmageed, Z.Y. Exosomes are the driving force in preparing the soil for the metastatic seeds: Lessons from the prostate cancer. Cells 2020, 9, 564. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  285. Melnik, B.C.; John, S.M.; Weiskirchen, R.; Schmitz, G. The endocrine and epigenetic impact of persistent cow milk consumption on prostate carcinogenesis. J. Transl. Genet. Genom. 2022, 6, 1–45. [Google Scholar] [CrossRef]
  286. Caner, V.; Cetin, G.O.; Hacioglu, S.; Baris, I.C.; Tepeli, E.; Turk, N.S.; Bagci, G.; Yararbas, K.; Cagliyan, G. The miRNA content of circulating exosomes in DLBCL patients and in vitro influence of DLBCL-derived exosomes on miRNA expression of healthy B-cells from peripheral blood. Cancer Biomark. 2021, 32, 519–529. [Google Scholar] [CrossRef]
  287. Feng, Y.; Zhong, M.; Zeng, S.; Wang, L.; Liu, P.; Xiao, X.; Liu, Y. Exosome-derived miRNAs as predictive biomarkers for diffuse large B-cell lymphoma chemotherapy resistance. Epigenomics 2019, 11, 35–51. [Google Scholar] [CrossRef] [Green Version]
  288. Zhang, L.; Zhou, S.; Zhou, T.; Li, X.; Tang, J. Potential of the tumor-derived extracellular vesicles carrying the miR-125b-5p target TNFAIP3 in reducing the sensitivity of diffuse large B cell lymphoma to rituximab. Int. J. Oncol. 2021, 58, 31. [Google Scholar] [CrossRef]
  289. Pujari, R.; Hunte, R.; Khan, W.N.; Shembade, N. A20-mediated negative regulation of canonical NF-κB signaling pathway. Immunol. Res. 2013, 57, 166–171. [Google Scholar] [CrossRef]
  290. Zetoune, F.S.; Murthy, A.R.; Shao, Z.; Hlaing, T.; Zeidler, M.G.; Li, Y.; Vincenz, C. A20 inhibits NF-kappa B activation downstream of multiple Map3 kinases and interacts with the I kappa B signalosome. Cytokine 2001, 15, 282–298. [Google Scholar] [CrossRef]
  291. Bu, R.; Bavi, P.; Abubaker, J.; Jehan, Z.; Al-Haqawi, W.; Ajarim, D.; Al-Dayel, F.; Uddin, S.; Al-Kuraya, K.S. Role of nuclear factor-κB regulators TNFAIP3 and CARD11 in Middle Eastern diffuse large B-cell lymphoma. Leuk. Lymphoma 2012, 53, 1971–1977. [Google Scholar] [CrossRef]
  292. Gamboa-Cedeño, A.M.; Castillo, M.; Xiao, W.; Waldmann, T.A.; Ranuncolo, S.M. Alternative and canonical NF-kB pathways DNA-binding hierarchies networks define Hodgkin lymphoma and Non-Hodgkin diffuse large B Cell lymphoma respectively. J. Cancer Res. Clin. Oncol. 2019, 145, 1437–1448. [Google Scholar] [CrossRef]
  293. Wenzl, K.; Manske, M.K.; Sarangi, V.; Asmann, Y.W.; Greipp, P.T.; Schoon, H.R.; Braggio, E.; Maurer, M.J.; Feldman, A.L.; Witzig, T.E.; et al. Loss of TNFAIP3 enhances MYD88L265P-driven signaling in non-Hodgkin lymphoma. Blood Cancer J. 2018, 8, 97. [Google Scholar] [CrossRef] [Green Version]
  294. Shaffer, A.L.; Lin, K.I.; Kuo, T.C.; Yu, X.; Hurt, E.M.; Rosenwald, A.; Giltnane, J.M.; Yang, L.; Zhao, H.; Calame, K.; et al. Blimp-1 orchestrates plasma cell differentiation by extinguishing the mature B cell gene expression program. Immunity 2002, 17, 51–62. [Google Scholar] [CrossRef] [Green Version]
  295. Nadeau, S.; Martins, G.A. Conserved and unique functions of Blimp1 in immune cells. Front. Immunol. 2022, 12, 805260. [Google Scholar] [CrossRef]
  296. Dent, A.L.; Shaffer, A.L.; Yu, X.; Allman, D.; Staudt, L.M. Control of inflammation, cytokine expression, and germinal center formation by BCL-6. Science 1997, 276, 589–592. [Google Scholar] [CrossRef]
  297. Ye, B.H.; Cattoretti, G.; Shen, Q.; Zhang, J.; Hawe, N.; de Waard, R.; Leung, C.; Nouri-Shirazi, M.; Orazi, A.; Chaganti, R.S.; et al. The BCL-6 proto-oncogene controls germinal-centre formation and Th2-type inflammation. Nat. Genet. 1997, 16, 161–170. [Google Scholar] [CrossRef] [PubMed]
  298. Fukuda, T.; Yoshida, T.; Okada, S.; Hatano, M.; Miki, T.; Ishibashi, K.; Okabe, S.; Koseki, H.; Hirosawa, S.; Taniguchi, M.; et al. Disruption of the Bcl6 gene results in an impaired germinal center formation. J. Exp. Med. 1997, 186, 439–448. [Google Scholar] [CrossRef] [PubMed]
  299. Ci, W.; Polo, J.M.; Cerchietti, L.; Shaknovich, R.; Wang, L.; Yang, S.N.; Ye, K.; Farinha, P.; Horsman, D.E.; Gascoyne, R.D.; et al. The BCL6 transcriptional program features repression of multiple oncogenes in primary B cells and is deregulated in DLBCL. Blood 2009, 113, 5536–5548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  300. Parekh, S.; Privé, G.; Melnick, A. Therapeutic targeting of the BCL6 oncogene for diffuse large B-cell lymphomas. Leuk. Lymphoma 2008, 49, 874–882. [Google Scholar] [CrossRef] [Green Version]
  301. Parekh, S.; Polo, J.M.; Shaknovich, R.; Juszczynski, P.; Lev, P.; Ranuncolo, S.M.; Yin, Y.; Klein, U.; Cattoretti, G.; Dalla Favera, R.; et al. BCL6 programs lymphoma cells for survival and differentiation through distinct biochemical mechanisms. Blood 2007, 110, 2067–2074. [Google Scholar] [CrossRef] [Green Version]
  302. Hatzi, K.; Melnick, A. Breaking bad in the germinal center: How deregulation of BCL6 contributes to lymphomagenesis. Trends Mol. Med. 2014, 20, 343–352. [Google Scholar] [CrossRef] [Green Version]
  303. Phan, R.T.; Dalla-Favera, R. The BCL6 proto-oncogene suppresses p53 expression in germinal-centre B cells. Nature 2004, 432, 635–639. [Google Scholar] [CrossRef]
  304. Reljic, R.; Wagner, S.D.; Peakman, L.J.; Fearon, D.T. Suppression of signal transducer and activator of transcription 3-dependent B lymphocyte terminal differentiation by BCL-6. J. Exp. Med. 2000, 192, 1841–1848. [Google Scholar]
  305. Xu-Monette, Z.Y.; Wu, L.; Visco, C.; Tai, Y.C.; Tzankov, A.; Liu, W.M.; Montes-Moreno, S.; Dybkaer, K.; Chiu, A.; Orazi, A.; et al. Mutational profile and prognostic significance of TP53 in diffuse large B-cell lymphoma patients treated with R-CHOP: Report from an International DLBCL Rituximab-CHOP Consortium Program Study. Blood 2012, 120, 3986–3996. [Google Scholar] [CrossRef]
  306. Pascual, M.; Mena-Varas, M.; Robles, E.F.; Garcia-Barchino, M.J.; Panizo, C.; Hervas-Stubbs, S.; Alignani, D.; Sagardoy, A.; Martinez-Ferrandis, J.I.; Bunting, K.L.; et al. PD-1/PD-L1 immune checkpoint and p53 loss facilitate tumor progression in activated B-cell diffuse large B-cell lymphomas. Blood 2019, 133, 2401–2412. [Google Scholar] [CrossRef]
  307. Pasqualucci, L.; Compagno, M.; Houldsworth, J.; Monti, S.; Grunn, A.; Nandula, S.V.; Aster, J.C.; Murty, V.V.; Shipp, M.A.; Dalla-Favera, R. Inactivation of the PRDM1/BLIMP1 gene in diffuse large B cell lymphoma. J. Exp. Med. 2006, 203, 311–317. [Google Scholar] [CrossRef] [Green Version]
  308. Tam, W.; Gomez, M.; Chadburn, A.; Lee, J.W.; Chan, W.C.; Knowles, D.M. Mutational analysis of PRDM1 indicates a tumor-suppressor role in diffuse large B-cell lymphomas. Blood 2006, 107, 4090–4100. [Google Scholar] [CrossRef]
  309. Kallies, A.; Nutt, S.L. Terminal differentiation of lymphocytes depends on Blimp-1. Curr. Opin. Immunol. 2007, 19, 156–162. [Google Scholar] [CrossRef]
  310. Nutt, S.L.; Fairfax, K.A.; Kallies, A. BLIMP1 guides the fate of effector B and T cells. Nat. Rev. Immunol. 2007, 7, 923–927. [Google Scholar] [CrossRef]
  311. Lossos, I.S. BLIMP1 against lymphoma: The verdict is reached. Cancer Cell 2010, 18, 537–539. [Google Scholar] [CrossRef] [Green Version]
  312. Hangaishi, A.; Kurokawa, M. Blimp-1 is a tumor suppressor gene in lymphoid malignancies. Int. J. Hematol. 2010, 91, 46–53. [Google Scholar] [CrossRef]
  313. Mandelbaum, J.; Bhagat, G.; Tang, H.; Mo, T.; Brahmachary, M.; Shen, Q.; Chadburn, A.; Rajewsky, K.; Tarakhovsky, A.; Pasqualucci, L.; et al. BLIMP1 is a tumor suppressor gene frequently disrupted in activated B cell-like diffuse large B cell lymphoma. Cancer Cell 2010, 18, 568–579. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  314. Calado, D.P.; Zhang, B.; Srinivasan, L.; Sasaki, Y.; Seagal, J.; Unitt, C.; Rodig, S.; Kutok, J.; Tarakhovsky, A.; Schmidt-Supprian, M.; et al. Constitutive canonical NF-κB activation cooperates with disruption of BLIMP1 in the pathogenesis of activated B cell-like diffuse large cell lymphoma. Cancer Cell 2010, 18, 580–589. [Google Scholar] [CrossRef] [Green Version]
  315. Boi, M.; Zucca, E.; Inghirami, G.; Bertoni, F. PRDM1/BLIMP1: A tumor suppressor gene in B and T cell lymphomas. Leuk. Lymphoma 2015, 56, 1223–1228. [Google Scholar] [CrossRef] [PubMed]
  316. Nong, L.; Zheng, Y.; Li, X.; Li, D.; Liang, L.; Wang, W.; Li, T. The genetic deletion and protein expression of PRDM1 and its clinical implications in diffuse large B cell lymphoma: A retrospective cohort study in China. Pathol. Res. Pract. 2022, 233, 153860. [Google Scholar] [CrossRef] [PubMed]
  317. Xia, Y.; Xu-Monette, Z.Y.; Tzankov, A.; Li, X.; Manyam, G.C.; Murty, V.; Bhagat, G.; Zhang, S.; Pasqualucci, L.; Visco, C.; et al. Loss of PRDM1/BLIMP-1 function contributes to poor prognosis of activated B-cell-like diffuse large B-cell lymphoma. Leukemia 2017, 31, 625–636. [Google Scholar] [CrossRef]
  318. Qing, K.; Jin, Z.; Xu, Z.; Wang, W.; Li, X.; Zhang, Y.; Wang, L.; Zhu, H.; Xiang, R.; Wu, S.; et al. Dysregulated MDR1 by PRDM1/Blimp1 is involved in the doxorubicin resistance of non-germinal center B-cell-like diffuse large B-cell lymphoma. Chemotherapy 2022, 67, 12–23. [Google Scholar] [CrossRef]
  319. Lin, Y.; Wong, K.; Calame, K. Repression of c-myc transcription by Blimp-1, an inducer of terminal B cell differentiation. Science 1997, 276, 596–599. [Google Scholar] [CrossRef]
  320. Zhang, X.Y.; Ma, Z.P.; Cui, W.L.; Pang, X.L.; Chen, R.; Wang, L.; Zhang, W.; Li, X.X. Impact of PRDM1 gene inactivation on C-MYC regulation in diffuse large B-cell lymphoma. Zhonghua Bing Li Xue Za Zhi 2018, 47, 25–31. [Google Scholar]
  321. Asirvatham, A.J.; Gregorie, C.J.; Hu, Z.; Magner, J.; Tomasi, T.B. MicroRNA targets in immune genes and the Dicer/Argonaute and ARE machinery components. Mol. Immunol. 2008, 45, 1995–2006. [Google Scholar] [CrossRef] [Green Version]
  322. Ma, J.; Nie, K.; Redmond, D.; Liu, Y.; Elemento, O.; Knowles, D.M.; Tam, W. EBV-miR-BHRF1-2 targets PRDM1/Blimp1: Potential role in EBV lymphomagenesis. Leukemia 2016, 30, 594–604. [Google Scholar] [CrossRef] [Green Version]
  323. Alsaadi, M.; Khan, M.Y.; Dalhat, M.H.; Bahashwan, S.; Khan, M.U.; Albar, A.; Almehdar, H.; Qadri, I. Dysregulation of miRNAs in DLBCL: Causative factor for pathogenesis, diagnosis and prognosis. Diagnostics 2021, 11, 1739. [Google Scholar] [CrossRef]
  324. Nie, K.; Gomez, M.; Landgraf, P.; Garcia, J.F.; Liu, Y.; Tan, L.H.; Chadburn, A.; Tuschl, T.; Knowles, D.M.; Tam, W. MicroRNA-mediated down-regulation of PRDM1/Blimp-1 in Hodgkin/Reed-Sternberg cells: A potential pathogenetic lesion in Hodgkin lymphomas. Am. J. Pathol. 2008, 173, 242–252. [Google Scholar] [CrossRef] [Green Version]
  325. Nie, K.; Zhang, T.; Allawi, H.; Gomez, M.; Liu, Y.; Chadburn, A.; Wang, Y.L.; Knowles, D.M.; Tam, W. Epigenetic down-regulation of the tumor suppressor gene PRDM1/Blimp-1 in diffuse large B cell lymphomas: A potential role of the microRNA let-7. Am. J. Pathol. 2010, 177, 1470–1479. [Google Scholar] [CrossRef]
  326. Malumbres, R.; Sarosiek, K.A.; Cubedo, E.; Ruiz, J.W.; Jiang, X.; Gascoyne, R.D.; Tibshirani, R.; Lossos, I.S. Differentiation stage-specific expression of microRNAs in B lymphocytes and diffuse large B-cell lymphomas. Blood 2009, 113, 3754–3764. [Google Scholar] [CrossRef] [Green Version]
  327. Lu, T.X.; Young, K.H.; Xu, W.; Li, J.Y. TP53 dysfunction in diffuse large B-cell lymphoma. Crit. Rev. Oncol. Hematol. 2016, 97, 47–55. [Google Scholar] [CrossRef]
  328. Le, M.T.; Shyh-Chang, N.; Khaw, S.L.; Chin, L.; The, C.; Tay, J.; O’Day, E.; Korzh, V.; Yang, H.; Lal, A.; et al. Conserved regulation of p53 network dosage by microRNA-125b occurs through evolving miRNA-target gene pairs. PLoS Genet. 2011, 7, e1002242. [Google Scholar] [CrossRef] [Green Version]
  329. Kumar, M.; Lu, Z.; Takwi, A.A.; Chen, W.; Callander, N.S.; Ramos, K.S.; Young, K.H.; Li, Y. Negative regulation of the tumor suppressor p53 gene by microRNAs. Oncogene 2011, 30, 843–853. [Google Scholar] [CrossRef] [Green Version]
  330. Li, Y.; Gordon, M.W.; Xu-Monette, Z.Y.; Visco, C.; Tzankov, A.; Zou, D.; Qiu, L.; Montes-Moreno, S.; Dybkaer, K.; Orazi, A.; et al. Single nucleotide variation in the TP53 3’ untranslated region in diffuse large B-cell lymphoma treated with rituximab-CHOP: A report from the International DLBCL Rituximab-CHOP Consortium Program. Blood 2013, 121, 4529–4540. [Google Scholar] [CrossRef] [Green Version]
  331. Manfè, V.; Biskup, E.; Willumsgaard, A.; Skov, A.G.; Palmieri, D.; Gasparini, P.; Laganá, A.; Woetmann, A.; Ødum, N.; Croce, C.M.; et al. cMyc/miR-125b-5p signalling determines sensitivity to bortezomib in preclinical model of cutaneous T-cell lymphomas. PLoS ONE 2013, 8, e59390. [Google Scholar] [CrossRef]
  332. Lawrie, C.H.; Gal, S.; Dunlop, H.M.; Pushkaran, B.; Liggins, A.P.; Pulford, K.; Banham, A.H.; Pezzella, F.; Boultwood, J.; Wainscoat, J.S.; et al. Detection of elevated levels of tumour-associated microRNAs in serum of patients with diffuse large B-cell lymphoma. Br. J. Haematol. 2008, 141, 672–675. [Google Scholar] [CrossRef]
  333. Chen, W.; Wang, H.; Chen, H.; Liu, S.; Lu, H.; Kong, D.; Huang, X.; Kong, Q.; Lu, Z. Clinical significance and detection of microRNA-21 in serum of patients with diffuse large B-cell lymphoma in Chinese population. Eur. J. Haematol. 2014, 92, 407–412. [Google Scholar] [CrossRef] [PubMed]
  334. Inada, K.; Okoshi, Y.; Cho, Y.; Saito, H.; Iijima, T.; Hori, M.; Kojima, H. Availability of circulating microRNAs as a biomarker for early diagnosis of diffuse large B-cell lymphoma. Open J. Blood Dis. 2015, 5, 48–58. [Google Scholar] [CrossRef] [Green Version]
  335. Li, J.; Fu, R.; Yang, L.; Tu, W. miR-21 expression predicts prognosis in diffuse large B-cell lymphoma. Int. J. Clin. Exp. Pathol. 2015, 8, 15019–15024. [Google Scholar] [PubMed]
  336. Larrabeiti-Etxebarria, A.; Lopez-Santillan, M.; Santos-Zorrozua, B.; Lopez-Lopez, E.; Garcia-Orad, A. Systematic review of the potential of microRNAs in diffuse large B cell lymphoma. Cancers 2019, 11, 144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  337. Lopez-Santillan, M.; Larrabeiti-Etxebarria, A.; Arzuaga-Mendez, J.; Lopez-Lopez, E.; Garcia-Orad, A. Circulating miRNAs as biomarkers in diffuse large B-cell lymphoma: A systematic review. Oncotarget 2018, 9, 22850–22861. [Google Scholar] [CrossRef] [Green Version]
  338. Abou Elnour, E.S.; El Sayed, I.E.T.; Mohamed Abd Elbary, H.; Sohaib, A.; Amin Mohammed Atia, S.; El Sayed Ramadan Genena, S. Biochemical and clinical impacts of miR-150 and miR-21 expression levels in diffuse large B cell lymphoma. J. Immunoass. Immunochem. 2022, 43, 648–664. [Google Scholar] [CrossRef]
  339. Roehle, A.; Hoefig, K.P.; Repsilber, D.; Thorns, C.; Ziepert, M.; Wesche, K.O.; Thiere, M.; Loeffler, M.; Klapper, W.; Pfreundschuh, M.; et al. MicroRNA signatures characterize diffuse large B-cell lymphomas and follicular lymphomas. Br. J. Haematol. 2008, 142, 732–744. [Google Scholar]
  340. Lawrie, C.H.; Soneji, S.; Marafioti, T.; Cooper, C.D.; Palazzo, S.; Paterson, J.C.; Cattan, H.; Enver, T.; Mager, R.; Boultwood, J.; et al. MicroRNA expression distinguishes between germinal center B cell-like and activated B cell-like subtypes of diffuse large B cell lymphoma. Int. J. Cancer 2007, 121, 1156–1161. [Google Scholar] [CrossRef]
  341. Musilova, K.; Mraz, M. MicroRNAs in B-cell lymphomas: How a complex biology gets more complex. Leukemia 2015, 29, 1004–1017. [Google Scholar] [CrossRef]
  342. Barnes, N.A.; Stephenson, S.; Cocco, M.; Tooze, R.M.; Doody, G.M. BLIMP-1 and STAT3 counterregulate microRNA-21 during plasma cell differentiation. J. Immunol. 2012, 189, 253–260. [Google Scholar] [CrossRef] [Green Version]
  343. Munch-Petersen, H.D.; Ralfkiaer, U.; Sjö, L.D.; Hother, C.; Asmar, F.; Nielsen, B.S.; Brown, P.; Ralfkiaer, E.; Grønbæk, K. Differential expression of miR-155 and miR-21 in tumor and stroma cells in diffuse large B-cell lymphoma. Appl. Immunohistochem. Mol. Morphol. 2015, 23, 188–195. [Google Scholar] [CrossRef]
  344. Cao, D.; Cao, X.; Jiang, Y.; Xu, J.; Zheng, Y.; Kang, D.; Xu, C. Circulating exosomal microRNAs as diagnostic and prognostic biomarkers in patients with diffuse large B-cell lymphoma. Hematol. Oncol. 2022, 40, 172–180. [Google Scholar] [CrossRef]
  345. Li, S.; He, X.; Gan, Y.; Zhang, J.; Gao, F.; Lin, L.; Qiu, X.; Yu, T.; Zhang, X.; Chen, P.; et al. Targeting miR-21 with NL101 blocks c-Myc/Mxd1 loop and inhibits the growth of B cell lymphoma. Theranostics 2021, 11, 3439–3451. [Google Scholar] [CrossRef]
  346. Chen, L.; Zhan, C.Z.; Wang, T.; You, H.; Yao, R. Curcumin inhibits the proliferation, migration, invasion, and apoptosis of diffuse large B-cell lymphoma cell line by regulating miR-21/VHL Axis. Yonsei Med. J. 2020, 61, 20–29. [Google Scholar] [CrossRef]
  347. Chen, J.; Xu, T.; Chen, C. The critical roles of miR-21 in anti-cancer effects of curcumin. Ann. Transl. Med. 2015, 3, 330. [Google Scholar]
  348. Choudhury, S.N.; Li, Y. miR-21 and let-7 in the Ras and NF-κB pathways. Microrna 2012, 1, 65–69. [Google Scholar] [CrossRef]
  349. Bai, D.; Ueno, L.; Vogt, P.K. Akt-mediated regulation of NFkappaB and the essentialness of NFkappaB for the oncogenicity of PI3K and Akt. Int. J. Cancer 2009, 125, 2863–2870. [Google Scholar] [CrossRef] [Green Version]
  350. Go, H.; Jang, J.Y.; Kim, P.J.; Kim, Y.G.; Nam, S.J.; Paik, J.H.; Kim, T.M.; Heo, D.S.; Kim, C.W.; Jeon, Y.K. MicroRNA-21 plays an oncogenic role by targeting FOXO1 and activating the PI3K/AKT pathway in diffuse large B-cell lymphoma. Oncotarget 2015, 6, 15035–15049. [Google Scholar] [CrossRef] [Green Version]
  351. Song, J.; Shao, Q.; Li, C.; Liu, H.; Li, J.; Wang, Y.; Song, W.; Li, L.; Wang, G.; Shao, Z.; et al. Effects of microRNA-21 on apoptosis by regulating the expression of PTEN in diffuse large B-cell lymphoma. Medicine 2017, 96, e7952. [Google Scholar] [CrossRef]
  352. Szydlowski, M.; Kiliszek, P.; Sewastianik, T.; Jablonska, E.; Bialopiotrowicz, E.; Gorniak, P.; Polak, A.; Markowicz, S.; Nowak, E.; Grygorowicz, M.A.; et al. FOXO1 activation is an effector of SYK and AKT inhibition in tonic BCR signal-dependent diffuse large B-cell lymphomas. Blood 2016, 127, 739–748. [Google Scholar] [CrossRef] [Green Version]
  353. Bouvy, C.; Wannez, A.; George, F.; Graux, C.; Chatelain, C.; Dogné, J.M. Circulating microRNAs as biomarkers in diffuse large B-cell lymphoma: A pilot prospective longitudinal clinical study. Biomark. Cancer 2018, 10, 1179299X18781095. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  354. Gu, L.; Song, G.; Chen, L.; Nie, Z.; He, B.; Pan, Y.; Xu, Y.; Li, R.; Gao, T.; Cho, W.C.; et al. Inhibition of miR-21 induces biological and behavioral alterations in diffuse large B-cell lymphoma. Acta Haematol. 2013, 130, 87–94. [Google Scholar] [CrossRef] [PubMed]
  355. Zheng, Z.; Li, X.; Zhu, Y.; Gu, W.; Xie, X.; Jiang, J. Prognostic significance of miRNA in patients with diffuse large B-cell lymphoma: A meta-analysis. Cell. Physiol. Biochem. 2016, 39, 1891–1904. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  356. Sun, R.; Zheng, Z.; Wang, L.; Cheng, S.; Shi, Q.; Qu, B.; Fu, D.; Leboeuf, C.; Zhao, Y.; Ye, J.; et al. A novel prognostic model based on four circulating miRNA in diffuse large B-cell lymphoma: Implications for the roles of MDSC and Th17 cells in lymphoma progression. Mol. Oncol. 2021, 15, 246–261. [Google Scholar] [CrossRef]
  357. Fuertes, T.; Ramiro, A.R.; de Yebenes, V.G. miRNA-based therapies in B cell non-Hodgkin lymphoma. Trends Immunol. 2020, 41, 932–947. [Google Scholar] [CrossRef]
  358. Thai, T.H.; Calado, D.P.; Casola, S.; Ansel, K.M.; Xiao, C.; Xue, Y.; Murphy, A.; Frendewey, D.; Valenzuela, D.; Kutok, J.L.; et al. Regulation of the germinal center response by microRNA-155. Science 2007, 316, 604–608. [Google Scholar] [CrossRef]
  359. Elton, T.S.; Selemon, H.; Elton, S.M.; Parinandi, N.L. Regulation of the MIR155 host gene in physiological and pathological processes. Gene 2013, 532, 1–12. [Google Scholar] [CrossRef]
  360. Eis, P.S.; Tam, W.; Sun, L.; Chadburn, A.; Li, Z.; Gomez, M.F.; Lund, E.; Dahlberg, J.E. Accumulation of miR-155 and BIC RNA in human B cell lymphomas. Proc. Natl. Acad. Sci. USA 2005, 102, 3627–3632. [Google Scholar] [CrossRef] [Green Version]
  361. Anastasiadou, E.; Seto, A.G.; Beatty, X.; Hermreck, M.; Gilles, M.E.; Stroopinsky, D.; Pinter-Brown, L.C.; Pestano, L.; Marchese, C.; Avigan, D.; et al. Cobomarsen, an oligonucleotide inhibitor of miR-155, slows DLBCL tumor cell growth in vitro and in vivo. Clin. Cancer Res. 2021, 27, 1139–1149. [Google Scholar] [CrossRef]
  362. Zhong, H.; Xu, L.; Zhong, J.H.; Xiao, F.; Liu, Q.; Huang, H.H.; Chen, F.Y. Clinical and prognostic significance of miR-155 and miR-146a expression levels in formalin-fixed/paraffin-embedded tissue of patients with diffuse large B-cell lymphoma. Exp. Ther. Med. 2012, 3, 763–770. [Google Scholar] [CrossRef] [Green Version]
  363. Havranek, O.; Xu, J.; Köhrer, S.; Wang, Z.; Becker, L.; Comer, J.M.; Henderson, J.; Ma, W.; Man Chun Ma, J.; Westin, J.R.; et al. Tonic B-cell receptor signaling in diffuse large B-cell lymphoma. Blood 2017, 130, 995–1006. [Google Scholar] [CrossRef] [Green Version]
  364. Huang, X.; Shen, Y.; Liu, M.; Bi, C.; Jiang, C.; Iqbal, J.; McKeithan, T.W.; Chan, W.C.; Ding, S.J.; Fu, K. Quantitative proteomics reveals that miR-155 regulates the PI3K-AKT pathway in diffuse large B-cell lymphoma. Am. J. Pathol. 2012, 181, 26–33. [Google Scholar] [CrossRef] [Green Version]
  365. Fu, X.; Wen, H.; Jing, L.; Yang, Y.; Wang, W.; Liang, X.; Nan, K.; Yao, Y.; Tian, T. MicroRNA-155-5p promotes hepatocellular carcinoma progression by suppressing PTEN through the PI3K/Akt pathway. Cancer Sci. 2017, 108, 620–631. [Google Scholar] [CrossRef] [Green Version]
  366. Sun, J.F.; Zhang, D.; Gao, C.J.; Zhang, Y.W.; Dai, Q.S. Exosome-mediated miR-155 transfer contributes to hepatocellular carcinoma cell proliferation by targeting PTEN. Med. Sci. Monit. Basic Res. 2019, 25, 218–228. [Google Scholar] [CrossRef]
  367. Jabłońska, E.; Białopiotrowicz, E.; Szydłowski, M.; Prochorec-Sobieszek, M.; Juszczyński, P.; Szumera-Ciećkiewicz, A. DEPTOR is a microRNA-155 target regulating migration and cytokine production in diffuse large B-cell lymphoma cells. Exp. Hematol. 2020, 88, 56–67. [Google Scholar] [CrossRef]
  368. Morales-Martinez, M.; Lichtenstein, A.; Vega, M.I. Function of Deptor and its roles in hematological malignancies. Aging (Albany NY) 2021, 13, 1528–1564. [Google Scholar] [CrossRef]
  369. Peterson, T.R.; Laplante, M.; Thoreen, C.C.; Sancak, Y.; Kang, S.A.; Kuehl, W.M.; Gray, N.S.; Sabatini, D.M. DEPTOR is an mTOR inhibitor frequently overexpressed in multiple myeloma cells and required for their survival. Cell 2009, 137, 873–886. [Google Scholar] [CrossRef] [Green Version]
  370. Fu, W.; Hall, M.N. Regulation of mTORC2 signaling. Genes 2020, 11, 1045. [Google Scholar] [CrossRef]
  371. Huang, W.T.; Kuo, S.H.; Kuo, Y.C.; Lin, C.W. miR-155-regulated mTOR and Toll-like receptor 5 in gastric diffuse large B-cell lymphoma. Cancer Med. 2022, 11, 555–570. [Google Scholar] [CrossRef]
  372. O’Connell, R.M.; Chaudhuri, A.A.; Rao, D.S.; Baltimore, D. Inositol phosphatase SHIP1 is a primary target of miR-155. Proc. Natl. Acad. Sci. USA 2009, 106, 7113–7118. [Google Scholar] [CrossRef] [Green Version]
  373. Damen, J.E.; Liu, L.; Rosten, P.; Humphries, R.K.; Jefferson, A.B.; Majerus, P.W.; Krystal, G. The 145-kDa protein induced to associate with Shc by multiple cytokines is an inositol tetraphosphate and phosphatidylinositol 3,4,5-triphosphate 5-phosphatase. Proc. Natl. Acad. Sci. USA 1996, 93, 1689–1693. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  374. Brauweiler, A.; Tamir, I.; Dal Porto, J.; Benschop, R.J.; Helgason, C.D.; Humphries, R.K.; Freed, J.H.; Cambier, J.C. Differential regulation of B cell development, activation, and death by the src homology 2 domain-containing 5’ inositol phosphatase (SHIP). J. Exp. Med. 2000, 191, 1545–1554. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  375. Pedersen, I.M.; Otero, D.; Kao, E.; Miletic, A.V.; Hother, C.; Ralfkiaer, E.; Rickert, R.C.; Gronbaek, K.; David, M. Onco-miR-155 targets SHIP1 to promote TNFalpha-dependent growth of B cell lymphomas. EMBO Mol. Med. 2009, 1, 288–295. [Google Scholar] [CrossRef] [PubMed]
  376. Lemm, E.A.; Valle-Argos, B.; Smith, L.D.; Richter, J.; Gebreselassie, Y.; Carter, M.J.; Karolova, J.; Svaton, M.; Helman, K.; Weston-Bell, N.J.; et al. Preclinical evaluation of a novel SHIP1 phosphatase activator for inhibition of PI3K signaling in malignant B cells. Clin. Cancer Res. 2020, 26, 1700–1711. [Google Scholar] [CrossRef]
  377. Ahmadvand, M.; Eskandari, M.; Pashaiefar, H.; Yaghmaie, M.; Manoochehrabadi, S.; Khakpour, G.; Sheikhsaran, F.; Montazer Zohour, M. Over expression of circulating miR-155 predicts prognosis in diffuse large B-cell lymphoma. Leuk. Res. 2018, 70, 45–48. [Google Scholar] [CrossRef]
  378. Zare, N.; Haghjooy Javanmard, S.; Mehrzad, V.; Eskandari, N.; Kefayat, A. Evaluation of exosomal miR-155, let-7g and let-7i levels as a potential noninvasive biomarker among refractory/relapsed patients, responsive patients and patients receiving R-CHOP. Leuk. Lymphoma 2019, 60, 1877–1889. [Google Scholar] [CrossRef]
  379. Seda, V.; Mraz, M. B-cell receptor signalling and its crosstalk with other pathways in normal and malignant cells. Eur. J. Haematol. 2015, 94, 193–205. [Google Scholar] [CrossRef]
  380. Gonzalez-Martin, A.; Adams, B.D.; Lai, M.; Shepherd, J.; Salvador-Bernaldez, M.; Salvador, J.M.; Lu, J.; Nemazee, D.; Xiao, C. The microRNA miR-148a functions as a critical regulator of B cell tolerance and autoimmunity. Nat. Immunol. 2016, 17, 433–440. [Google Scholar] [CrossRef]
  381. Liebermann, D.A.; Hoffman, B. Gadd45 in stress signaling. J. Mol. Signal. 2008, 3, 15. [Google Scholar] [CrossRef] [Green Version]
  382. Hollander, M.C.; Fornace, A.J., Jr. Genomic instability, centrosome amplification, cell cycle checkpoints and Gadd45a. Oncogene 2002, 21, 6228–6233. [Google Scholar] [CrossRef] [Green Version]
  383. Bhalla, S.; Evens, A.M.; Dai, B.; Prachand, S.; Gordon, L.I.; Gartenhaus, R.B. The novel anti-MEK small molecule AZD6244 induces BIM-dependent and AKT-independent apoptosis in diffuse large B-cell lymphoma. Blood 2011, 118, 1052–1061. [Google Scholar] [CrossRef] [Green Version]
  384. Porstner, M.; Winkelmann, R.; Daum, P.; Schmid, J.; Pracht, K.; Côrte-Real, J.; Schreiber, S.; Haftmann, C.; Brandl, A.; Mashreghi, M.F.; et al. miR-148a promotes plasma cell differentiation and targets the germinal center transcription factors Mitf and Bach2. Eur. J. Immunol. 2015, 45, 1206–1215. [Google Scholar] [CrossRef]
  385. Ichikawa, S.; Fukuhara, N.; Katsushima, H.; Takahashi, T.; Yamamoto, J.; Yokoyama, H.; Sasaki, O.; Fukuhara, O.; Nomura, J.; Ishizawa, K.; et al. Association between BACH2 expression and clinical prognosis in diffuse large B-cell lymphoma. Cancer Sci. 2014, 105, 437–444. [Google Scholar] [CrossRef]
  386. Pracht, K.; Meinzinger, J.; Schulz, S.R.; Daum, P.; Côrte-Real, J.; Hauke, M.; Roth, E.; Kindermann, D.; Mielenz, D.; Schuh, W.; et al. miR-148a controls metabolic programming and survival of mature CD19-negative plasma cells in mice. Eur. J. Immunol. 2021, 51, 1089–1109. [Google Scholar] [CrossRef]
  387. Melnik, B.C.; Schmitz, G. Pasteurized non-fermented cow’s milk but not fermented milk is a promoter of mTORC1-driven aging and increased mortality. Ageing Res. Rev. 2021, 67, 101270. [Google Scholar] [CrossRef]
  388. International Dairy Federation. Heat Treatment of Milk—Overview, 001/2018-02. IDF Factsheet. 2018. Available online: http://www.ukidf.org/documents/Factsheet-001_2018_Heat-treatment-1-2_001.pdf (accessed on 20 January 2023).
  389. Kleinjan, M.; van Herwijnen, M.J.; Libregts, S.F.; van Neerven, R.J.; Feitsma, A.L.; Wauben, M.H. Regular industrial processing of bovine milk impacts the integrity and molecular composition of extracellular vesicles. J. Nutr. 2021, 151, 1416–1425. [Google Scholar] [CrossRef]
  390. Zhang, Y.; Xu, Q.; Hou, J.; Huang, G.; Zhao, S.; Zheng, N.; Wang, J. Loss of bioactive microRNAs in cow’s milk by ultra-high-temperature treatment but not by pasteurization treatment. J. Sci. Food Agric. 2022, 102, 2676–2685. [Google Scholar] [CrossRef]
  391. Ngu, A.; Wang, S.; Wang, H.; Khanam, A.; Zempleni, J. Milk exosomes in nutrition and drug delivery. Am. J. Physiol. Cell Physiol. 2022, 322, C865–C874. [Google Scholar] [CrossRef]
  392. Benmoussa, A.; Lee, C.H.; Laffont, B.; Savard, P.; Laugier, J.; Boilard, E.; Gilbert, C.; Fliss, I.; Provost, P. Commercial dairy cow milk microRNAs resist digestion under simulated gastrointestinal tract conditions. J. Nutr. 2016, 146, 2206–2215. [Google Scholar] [CrossRef] [Green Version]
  393. Rani, P.; Vashisht, M.; Golla, N.; Vashisht, M.; Shandilya, S. Milk miRNAs encapsulated in exosomes are stable to human digestion and permeable to intestinal barrier in vitro. J. Funct. Foods 2017, 34, 431–439. [Google Scholar] [CrossRef]
  394. Wolf, T.; Baier, S.R.; Zempleni, J. The intestinal transport of bovine milk exosomes is mediated by endocytosis in human colon carcinoma Caco-2 cells and rat small intestinal IEC-6 cells. J. Nutr. 2015, 145, 2201–2206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  395. Kusuma, R.J.; Manca, S.; Friemel, T.; Sukreet, S.; Nguyen, C.; Zempleni, J. Human vascular endothelial cells transport foreign exosomes from cow’s milk by endocytosis. Am. J. Physiol. Cell Physiol. 2016, 310, C800–C807. [Google Scholar] [CrossRef] [Green Version]
  396. González-Sarrías, A.; Iglesias-Aguirre, C.E.; Cortés-Martín, A.; Vallejo, F.; Cattivelli, A.; Del Pozo-Acebo, L.; Del Saz, A.; López de Las Hazas, M.C.; Dávalos, A.; Espín, J.C. Milk-derived exosomes as nanocarriers to deliver curcumin and resveratrol in breast tissue and enhance their anticancer activity. Int. J. Mol. Sci. 2022, 23, 2860. [Google Scholar] [CrossRef] [PubMed]
  397. Wang, L.; Sadri, M.; Giraud, D.; Zempleni, J. RNase H2-dependent polymerase chain reaction and elimination of confounders in sample collection, storage, and analysis strengthen evidence that microRNAs in bovine milk are bioavailable in humans. J. Nutr. 2018, 148, 153–159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  398. Lin, D.; Chen, T.; Xie, M.; Li, M.; Zeng, B.; Sun, R.; Zhu, Y.; Ye, D.; Wu, J.; Sun, J.; et al. Oral administration of bovine and porcine milk exosome alter miRNAs profiles in piglet serum. Sci. Rep. 2020, 10, 6983. [Google Scholar] [CrossRef] [Green Version]
  399. Feng, X.; Chen, X.; Zheng, X.; Zhu, H.; Qi, Q.; Liu, S.; Zhang, H.; Che, J. Latest trend of milk derived exosomes: Cargos, functions, and applications. Front. Nutr. 2021, 8, 747294. [Google Scholar] [CrossRef]
  400. López de Las Hazas, M.C.; Del Pozo-Acebo, L.; Hansen, M.S.; Gil-Zamorano, J.; Mantilla-Escalante, D.C.; Gómez-Coronado, D.; Marín, F.; Garcia-Ruiz, A.; Rasmussen, J.T.; Dávalos, A. Dietary bovine milk miRNAs transported in extracellular vesicles are partially stable during GI digestion, are bioavailable and reach target tissues but need a minimum dose to impact on gene expression. Eur. J. Nutr. 2022, 61, 1043–1056. [Google Scholar] [CrossRef]
  401. Manca, S.; Upadhyaya, B.; Mutai, E.; Desaulniers, A.T.; Cederberg, R.A.; White, B.R.; Zempleni, J. Milk exosomes are bioavailable and distinct microRNA cargos have unique tissue distribution patterns. Sci. Rep. 2018, 8, 11321. [Google Scholar] [CrossRef] [Green Version]
  402. Betker, J.L.; Angle, B.M.; Graner, M.W.; Anchordoquy, T.J. The potential of exosomes from cow milk for oral delivery. J. Pharm. Sci. 2019, 108, 1496–1505. [Google Scholar] [CrossRef]
  403. Aguilar-Lozano, A.; Baier, S.; Grove, R.; Shu, J.; Giraud, D.; Leiferman, A.; Mercer, K.E.; Cui, J.; Badger, T.M.; Adamec, J.; et al. Concentrations of purine metabolites are elevated in fluids from adults and infants and in livers from mice fed diets depleted of bovine milk exosomes and their RNA cargos. J. Nutr. 2018, 148, 1886–1894. [Google Scholar] [CrossRef] [Green Version]
  404. Sadri, M.; Shu, J.; Kachman, S.D.; Cui, J.; Zempleni, J. Milk exosomes and miRNA cross the placenta and promote embryo survival in mice. Reproduction 2020, 160, 501–509. [Google Scholar] [CrossRef]
  405. Sanwlani, R.; Fonseka, P.; Chitti, S.V.; Mathivanan, S. Milk-derived extracellular vesicles in inter-organism, cross-species communication and drug delivery. Proteomes 2020, 8, 11. [Google Scholar] [CrossRef]
  406. Sanwlani, R.; Fonseka, P.; Mathivanan, S. Are dietary extracellular vesicles bioavailable and functional in consuming organisms? Subcell. Biochem. 2021, 97, 509–521. [Google Scholar]
  407. Askenase, P.W. Ancient evolutionary origin and properties of universally produced natural exosomes contribute to their therapeutic superiority compared to artificial nanoparticles. Int. J. Mol. Sci. 2021, 22, 1429. [Google Scholar] [CrossRef]
  408. Samuel, M.; Fonseka, P.; Sanwlani, R.; Gangoda, L.; Chee, S.H.; Keerthikumar, S.; Spurling, A.; Chitti, S.V.; Zanker, D.; Ang, C.S.; et al. Oral administration of bovine milk-derived extracellular vesicles induces senescence in the primary tumor but accelerates cancer metastasis. Nat. Commun. 2021, 12, 3950. [Google Scholar] [CrossRef]
  409. Baier, S.R.; Nguyen, C.; Xie, F.; Wood, J.R.; Zempleni, J. MicroRNAs are absorbed in biologically meaningful amounts from nutritionally relevant doses of cow milk and affect gene expression in peripheral blood mononuclear cells, HEK-293 kidney cell cultures, and mouse livers. J. Nutr. 2014, 144, 1495–1500. [Google Scholar] [CrossRef] [Green Version]
  410. Lässer, C.; Alikhani, V.S.; Ekström, K.; Eldh, M.; Paredes, P.T.; Bossios, A.; Sjöstrand, M.; Gabrielsson, S.; Lötvall, J.; Valadi, H. Human saliva, plasma and breast milk exosomes contain RNA: Uptake by macrophages. J. Transl. Med. 2011, 9, 9. [Google Scholar] [CrossRef] [Green Version]
  411. Izumi, H.; Tsuda, M.; Sato, Y.; Kosaka, N.; Ochiya, T.; Iwamoto, H.; Namba, K.; Takeda, Y. Bovine milk exosomes contain microRNA and mRNA and are taken up by human macrophages. J. Dairy Sci. 2015, 98, 2920–2933. [Google Scholar] [CrossRef] [Green Version]
  412. He, Y.; He, Z.; Leone, S.; Liu, S. Milk exosomes transfer oligosaccharides into macrophages to modulate immunity and attenuate adherent-invasive E. coli (AIEC) infection. Nutrients 2021, 13, 3198. [Google Scholar] [CrossRef]
  413. Admyre, C.; Johansson, S.M.; Qazi, K.R.; Filén, J.J.; Lahesmaa, R.; Norman, M.; Neve, E.P.; Scheynius, A.; Gabrielsson, S. Exosomes with immune modulatory features are present in human breast milk. J. Immunol. 2007, 179, 1969–1978. [Google Scholar] [CrossRef] [Green Version]
  414. Vashisht, M.; Rani, P.; Onteru, S.K.; Singh, D. Curcumin encapsulated in milk exosomes resists human digestion and possesses enhanced intestinal permeability in vitro. Appl. Biochem. Biotechnol. 2017, 183, 993–1007. [Google Scholar] [CrossRef]
  415. Salisbury, J.L.; Condeelis, J.S.; Satir, P. Role of coated vesicles, microfilaments, and calmodulin in receptor-mediated endocytosis by cultured B lymphoblastoid cells. J. Cell Biol. 1980, 87, 132–141. [Google Scholar] [CrossRef] [PubMed]
  416. Guagliardi, L.E.; Koppelman, B.; Blum, J.S.; Marks, M.S.; Cresswell, P.; Brodsky, F.M. Co-localization of molecules involved in antigen processing and presentation in an early endocytic compartment. Nature 1990, 343, 133–139. [Google Scholar] [CrossRef] [PubMed]
  417. Hoogeboom, R.; Tolar, P. Molecular mechanisms of B cell antigen gathering and endocytosis. Curr. Top. Microbiol. Immunol. 2016, 393, 45–63. [Google Scholar] [PubMed]
  418. Roberts, A.D.; Davenport, T.M.; Dickey, A.M.; Ahn, R.; Sochacki, K.A.; Taraska, J.W. Structurally distinct endocytic pathways for B cell receptors in B lymphocytes. Mol. Biol. Cell. 2020, 31, 2826–2840. [Google Scholar] [CrossRef]
  419. Malinova, D.; Wasim, L.; Newman, R.; Martínez-Riaño, A.; Engels, N.; Tolar, P. Endophilin A2 regulates B-cell endocytosis and is required for germinal center and humoral responses. EMBO Rep. 2021, 22, e51328. [Google Scholar] [CrossRef]
  420. McShane, A.N.; Malinova, D. The ins and outs of antigen uptake in B cells. Front. Immunol. 2022, 13, 892169. [Google Scholar] [CrossRef]
  421. Khanam, A.; Yu, J.; Zempleni, J. Class A scavenger receptor-1/2 facilitates the uptake of bovine milk exosomes in murine bone marrow-derived macrophages and C57BL/6J mice. Am. J. Physiol. Cell Physiol. 2021, 321, C607–C614. [Google Scholar] [CrossRef]
  422. Baos, S.; Cremades-Jimeno, L.; López-Ramos, M.; de Pedro, M.Á.; Uriarte, S.A.; Sastre, J.; González-Mangado, N.; Rodríguez-Nieto, M.J.; Peces-Barba, G.; Cárdaba, B. Expression of macrophage scavenger receptor (MSR1) in peripheral blood cells from patients with different respiratory diseases: Beyond monocytes. J. Clin. Med. 2022, 11, 1439. [Google Scholar] [CrossRef]
  423. Nam, S.J.; Kim, S.; Kwon, D.; Kim, H.; Kim, S.; Lee, E.; Kim, T.M.; Heo, D.S.; Park, S.H.; Lim, M.S.; et al. Prognostic implications of tumor-infiltrating macrophages, M2 macrophages, regulatory T-cells, and indoleamine 2,3-dioxygenase-positive cells in primary diffuse large B-cell lymphoma of the central nervous system. Oncoimmunology 2018, 7, e1442164. [Google Scholar] [CrossRef]
  424. van Bilsen, K.; Bastiaans, J.; Dik, W.A.; De Haas, E.F.; Baarsma, S.G.; Kuipers, R.W.; van Hagen, P.M. The neonatal Fc receptor is expressed by human lymphocytes. J. Transl. Med. 2010, 8 (Suppl. S1), P1. [Google Scholar] [CrossRef] [Green Version]
  425. Montoyo, H.P.; Vaccaro, C.; Hafner, M.; Ober, R.J.; Mueller, W.; Ward, E.S. Conditional deletion of the MHC class I-related receptor FcRn reveals the sites of IgG homeostasis in mice. Proc. Natl. Acad. Sci. USA 2009, 106, 2788–2793. [Google Scholar] [CrossRef] [Green Version]
  426. Mi, W.; Wanjie, S.; Lo, S.T.; Gan, Z.; Pickl-Herk, B.; Ober, R.J.; Ward, E.S. Targeting the neonatal fc receptor for antigen delivery using engineered fc fragments. J. Immunol. 2008, 181, 7550–7561. [Google Scholar] [CrossRef] [Green Version]
  427. Golan-Gerstl, R.; Elbaum Shiff, Y.; Moshayoff, V.; Schecter, D.; Leshkowitz, D.; Reif, S. Characterization and biological function of milk-derived miRNAs. Mol. Nutr. Food Res. 2017, 61, 1700009. [Google Scholar] [CrossRef]
  428. van Herwijnen, M.J.C.; Driedonks, T.A.P.; Snoek, B.L.; Kroon, A.M.T.; Kleinjan, M.; Jorritsma, R.; Pieterse, C.M.J.; Hoen, E.N.M.N.; Wauben, M.H.M. Abundantly present miRNAs in milk-derived extracellular vesicles are conserved between mammals. Front. Nutr. 2018, 5, 81. [Google Scholar] [CrossRef]
  429. Oh, S.; Park, M.R.; Son, S.J.; Kim, Y. Comparison of total RNA isolation methods for analysis of immune-related microRNAs in market milks. Korean J. Food Sci. Anim. Resour. 2015, 35, 459–465. [Google Scholar] [CrossRef] [Green Version]
  430. Benmoussa, A.; Laugier, J.; Beauparlant, C.J.; Lambert, M.; Droit, A.; Provost, P. Complexity of the microRNA transcriptome of cow milk and milk-derived extracellular vesicles isolated via differential ultracentrifugation. J. Dairy Sci. 2020, 103, 16–29. [Google Scholar] [CrossRef]
  431. Mecocci, S.; Pietrucci, D.; Milanesi, M.; Pascucci, L.; Filippi, S.; Rosato, V.; Chillemi, G.; Capomaccio, S.; Cappelli, K. Transcriptomic characterization of cow, donkey and goat milk extracellular vesicles reveals their anti-inflammatory and immunomodulatory potential. Int. J. Mol. Sci. 2021, 22, 12759. [Google Scholar] [CrossRef]
  432. Rashidi, M.; Bijari, S.; Khazaei, A.H.; Shojaei-Ghahrizjani, F.; Rezakhani, L. The role of milk-derived exosomes in the treatment of diseases. Front. Genet. 2022, 13, 1009338. [Google Scholar] [CrossRef]
  433. Rani, P.; Onteru, S.K.; Singh, D. Genome-wide profiling and analysis of microRNA expression in buffalo milk exosomes. Food Biosci. 2020, 38, 100769. [Google Scholar] [CrossRef]
  434. Chen, Z.; Xie, Y.; Luo, J.; Chen, T.; Xi, Q.; Zhang, Y.; Sun, J. Milk exosome-derived miRNAs from water buffalo are implicated in immune response and metabolism process. BMC Vet. Res. 2020, 16, 123. [Google Scholar] [CrossRef] [PubMed]
  435. Reif, S.; Atias, A.; Musseri, M.; Koroukhov, N.; Gerstl, R.G. Beneficial effects of milk-derived extracellular vesicles on liver fibrosis progression by inhibiting hepatic stellate cell activation. Nutrients 2022, 14, 4049. [Google Scholar] [CrossRef]
  436. Baddela, V.S.; Nayan, V.; Rani, P.; Onteru, S.K.; Singh, D. Physicochemical biomolecular insights into Buffalo milk-derived nanovesicles. Appl. Biochem. Biotechnol. 2016, 178, 544–557. [Google Scholar] [CrossRef] [PubMed]
  437. Schmitz, R.; Hansmann, M.L.; Bohle, V.; Martin-Subero, J.I.; Hartmann, S.; Mechtersheimer, G.; Klapper, W.; Vater, I.; Giefing, M.; Gesk, S.; et al. TNFAIP3 (A20) is a tumor suppressor gene in Hodgkin lymphoma and primary mediastinal B cell lymphoma. J. Exp. Med. 2009, 206, 981–989. [Google Scholar] [CrossRef] [Green Version]
  438. Kato, M.; Sanada, M.; Kato, I.; Sato, Y.; Takita, J.; Takeuchi, K.; Niwa, A.; Chen, Y.; Nakazaki, K.; Nomoto, J.; et al. Frequent inactivation of A20 in B-cell lymphomas. Nature 2009, 459, 712–716. [Google Scholar] [CrossRef] [PubMed]
  439. Heyninck, K.; De Valck, D.; Vanden Berghe, W.; Van Criekinge, W.; Contreras, R.; Fiers, W.; Haegeman, G.; Beyaert, R. The zinc finger protein A20 inhibits TNF-induced NF-kappaB-dependent gene expression by interfering with an RIP- or TRAF2-mediated transactivation signal and directly binds to a novel NF-kappaB-inhibiting protein ABIN. J. Cell Biol. 1999, 145, 1471–1482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  440. Wertz, I.E.; O’Rourke, K.M.; Zhou, H.; Eby, M.; Aravind, L.; Seshagiri, S.; Wu, P.; Wiesmann, C.; Baker, R.; Boone, D.L.; et al. De-ubiquitination and ubiquitin ligase domains of A20 downregulate NF-kappaB signalling. Nature 2004, 430, 694–699. [Google Scholar] [CrossRef]
  441. Shembade, N.; Ma, A.; Harhaj, E.W. Inhibition of NF-kappaB signaling by A20 through disruption of ubiquitin enzyme complexes. Science 2010, 327, 1135–1139. [Google Scholar] [CrossRef] [Green Version]
  442. Kim, S.W.; Ramasamy, K.; Bouamar, H.; Lin, A.P.; Jiang, D.; Aguiar, R.C. MicroRNAs miR-125a and miR-125b constitutively activate the NF-κB pathway by targeting the tumor necrosis factor alpha-induced protein 3 (TNFAIP3, A20). Proc. Natl. Acad. Sci. USA 2012, 109, 7865–7870. [Google Scholar] [CrossRef] [Green Version]
  443. Shome, S.; Jernigan, R.L.; Beitz, D.C.; Clark, S.; Testroet, E.D. Non-coding RNA in raw and commercially processed milk and putative targets related to growth and immune-response. BMC Genomics 2021, 22, 749. [Google Scholar] [CrossRef]
  444. Sun, Y.M.; Lin, K.Y.; Chen, Y.Q. Diverse functions of miR-125 family in different cell contexts. J. Hematol. Oncol. 2013, 6, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  445. Fromm, B.; Tosar, J.P.; Lu, Y.; Halushka, M.K.; Witwer, K.W. Human and cow have identical miR-21-5p and miR-30a-5p sequences, which are likely unsuited to study dietary uptake from cow milk. J. Nutr. 2018, 148, 1506–1507. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  446. Mutai, E.; Ramer-Tait, A.E.; Zempleni, J. MicroRNAs in bovine milk exosomes are bioavailable in humans but do not elicit a robust pro-inflammatory cytokine response. ExRNA 2020, 2, 2. [Google Scholar] [CrossRef] [Green Version]
  447. Chen, X.; Gao, C.; Li, H.; Huang, L.; Sun, Q.; Dong, Y.; Tian, C.; Gao, S.; Dong, H.; Guan, D.; et al. Identification and characterization of microRNAs in raw milk during different periods of lactation, commercial fluid, and powdered milk products. Cell Res. 2010, 20, 1128–1137. [Google Scholar] [CrossRef] [PubMed]
  448. Yu, S.; Zhao, Z.; Sun, L.; Li, P. Fermentation results in quantitative changes in milk-derived exosomes and different effects on cell growth and survival. J. Agric. Food Chem. 2017, 65, 1220–1228. [Google Scholar] [CrossRef]
  449. Ding, X.; Jing, N.; Shen, A.; Guo, F.; Song, Y.; Pan, M.; Ma, X.; Zhao, L.; Zhang, H.; Wu, L.; et al. MiR-21-5p in macrophage-derived extracellular vesicles affects podocyte pyroptosis in diabetic nephropathy by regulating A20. J. Endocrinol. Investig. 2021, 44, 1175–1184. [Google Scholar] [CrossRef]
  450. Xue, Z.; Xi, Q.; Liu, H.; Guo, X.; Zhang, J.; Zhang, Z.; Li, Y.; Yang, G.; Zhou, D.; Yang, H.; et al. miR-21 promotes NLRP3 inflammasome activation to mediate pyroptosis and endotoxic shock. Cell Death Dis. 2019, 10, 461. [Google Scholar] [CrossRef] [Green Version]
  451. Lei, B.X.; Liu, Z.H.; Li, Z.J.; Li, C.; Deng, Y.F. miR-21 induces cell proliferation and suppresses the chemosensitivity in glioblastoma cells via downregulation of FOXO1. Int. J. Clin. Exp. Med. 2014, 7, 2060–2066. [Google Scholar]
  452. Song, W.; Wang, L.; Wang, L.; Li, Q. Interplay of miR-21 and FoxO1 modulates growth of pancreatic ductal adenocarcinoma. Tumour Biol. 2015, 36, 4741–4745. [Google Scholar] [CrossRef]
  453. Song, W.; Li, Q.; Wang, L.; Wang, L. Modulation of FoxO1 expression by miR-21 to promote growth of pancreatic ductal adenocarcinoma. Cell. Physiol. Biochem. 2015, 35, 184–190. [Google Scholar] [CrossRef]
  454. Liu, H.Y.; Zhang, Y.Y.; Zhu, B.L.; Feng, F.Z.; Yan, H.; Zhang, H.Y.; Zhou, B. miR-21 regulates the proliferation and apoptosis of ovarian cancer cells through PTEN/PI3K/AKT. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 4149–4155. [Google Scholar]
  455. Wang, Z.; Zhou, H.; Cheng, F.; Zhang, Z.; Long, S. MiR-21 regulates epithelial-mesenchymal transition in intestinal fibrosis of Crohn’s disease by targeting PTEN/mTOR. Dig. Liver Dis. 2022, 54, 1358–1366. [Google Scholar] [CrossRef]
  456. Xie, X.; Qu, P.; Wu, H.; Liu, P.; Luo, J.; Chi, J.; Liu, X.; Chen, X.; Xu, C. Circulating exosomal miR-21 mediates HUVEC proliferation and migration through PTEN/PI3K/AKT in Crohn’s disease. Ann. Transl. Med. 2022, 10, 258. [Google Scholar] [CrossRef]
  457. Zhang, L.; Yu, L.; Liu, Y.; Wang, S.; Hou, Z.; Zhou, J. miR-21-5p promotes cell proliferation by targeting BCL11B in Thp-1 cells. Oncol. Lett. 2021, 21, 119. [Google Scholar] [CrossRef]
  458. Wakabayashi, Y.; Inoue, J.; Takahashi, Y.; Matsuki, A.; Kosugi-Okano, H.; Shinbo, T.; Mishima, Y.; Niwa, O.; Kominami, R. Homozygous deletions and point mutations of the Rit1/Bcl11b gene in gamma-ray induced mouse thymic lymphomas. Biochem. Biophys. Res. Commun. 2003, 301, 598–603. [Google Scholar] [CrossRef]
  459. Kominami, R. Role of the transcription factor Bcl11b in development and lymphomagenesis. Proc. Jpn. Acad. Ser. B Phys. Biol. Sci. 2012, 88, 72–87. [Google Scholar] [CrossRef] [Green Version]
  460. Thompson, P.A.; Eam, B.; Young, N.P.; Fish, S.; Chen, J.; Barrera, M.; Howard, H.; Sung, E.; Parra, A.; Staunton, J.; et al. Targeting oncogene mRNA translation in B-cell malignancies with eFT226, a potent and selective inhibitor of eIF4A. Mol. Cancer Ther. 2021, 20, 26–36. [Google Scholar] [CrossRef]
  461. Dorrello, N.V.; Peschiaroli, A.; Guardavaccaro, D.; Colburn, N.H.; Sherman, N.E.; Pagano, M. S6K1- and betaTRCP-mediated degradation of PDCD4 promotes protein translation and cell growth. Science 2006, 314, 467–471. [Google Scholar] [CrossRef]
  462. Yang, C.H.; Yue, J.; Pfeffer, S.R.; Fan, M.; Paulus, E.; Hosni-Ahmed, A.; Sims, M.; Qayyum, S.; Davidoff, A.M.; Handorf, C.R.; et al. MicroRNA-21 promotes glioblastoma tumorigenesis by down-regulating insulin-like growth factor-binding protein-3 (IGFBP3). J. Biol. Chem. 2014, 289, 25079–25087. [Google Scholar] [CrossRef] [Green Version]
  463. Satzger, I.; Mattern, A.; Kuettler, U.; Weinspach, D.; Niebuhr, M.; Kapp, A.; Gutzmer, R. microRNA-21 is upregulated in malignant melanoma and influences apoptosis of melanocytic cells. Exp. Dermatol. 2012, 21, 509–514. [Google Scholar] [CrossRef]
  464. Melnik, B.C. MiR-21: An environmental driver of malignant melanoma? J. Transl. Med. 2015, 13, 202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  465. Singh, A.; Singh, A.K.; Giri, R.; Kumar, D.; Sharma, R.; Valis, M.; Kuca, K.; Garg, N. The role of microRNA-21 in the onset and progression of cancer. Future Med. Chem. 2021, 13, 1885–1906. [Google Scholar] [CrossRef] [PubMed]
  466. Cioroianu, A.I.; Stinga, P.I.; Sticlaru, L.; Cioplea, M.D.; Nichita, L.; Popp, C.; Staniceanu, F. Tumor microenvironment in diffuse large B-cell lymphoma: Role and prognosis. Anal. Cell Pathol. 2019, 2019, 8586354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  467. Ng, W.L.; Ansell, S.M.; Mondello, P. Insights into the tumor microenvironment of B cell lymphoma. J. Exp. Clin. Cancer Res. 2022, 41, 362. [Google Scholar] [CrossRef] [PubMed]
  468. Opinto, G.; Vegliante, M.C.; Negri, A.; Skrypets, T.; Loseto, G.; Pileri, S.A.; Guarini, A.; Ciavarella, S. The tumor microenvironment of DLBCL in the computational era. Front. Oncol. 2020, 10, 351. [Google Scholar] [CrossRef] [Green Version]
  469. Shen, L.; Li, H.; Shi, Y.; Wang, D.; Gong, J.; Xun, J.; Zhou, S.; Xiang, R.; Tan, X. M2 tumour-associated macrophages contribute to tumour progression via legumain remodelling the extracellular matrix in diffuse large B cell lymphoma. Sci. Rep. 2016, 6, 30347. [Google Scholar] [CrossRef] [Green Version]
  470. Mitteldorf, C.; Berisha, A.; Pfaltz, M.C.; Broekaert, S.M.C.; Schön, M.P.; Kerl, K.; Kempf, W. Tumor microenvironment and checkpoint molecules in primary cutaneous diffuse large B-cell lymphoma-new therapeutic targets. Am. J. Surg. Pathol. 2017, 41, 998–1004. [Google Scholar] [CrossRef] [Green Version]
  471. Zhu, M.Y.; Liu, W.J.; Wang, H.; Wang, W.D.; Liu, N.W.; Lu, Y. NSE from diffuse large B-cell lymphoma cells regulates macrophage polarization. Cancer Manag. Res. 2019, 11, 4577–4595. [Google Scholar] [CrossRef] [Green Version]
  472. Huang, Y.H.; Cai, K.; Xu, P.P.; Wang, L.; Huang, C.X.; Fang, Y.; Cheng, S.; Sun, X.J.; Liu, F.; Huang, J.Y.; et al. CREBBP/EP300 mutations promoted tumor progression in diffuse large B-cell lymphoma through altering tumor-associated macrophage polarization via FBXW7-NOTCH-CCL2/CSF1 axis. Signal Transduct. Target Ther. 2021, 6, 10. [Google Scholar] [CrossRef]
  473. An, Y.; Yang, Q. MiR-21 modulates the polarization of macrophages and increases the effects of M2 macrophages on promoting the chemoresistance of ovarian cancer. Life Sci. 2020, 242, 117162. [Google Scholar] [CrossRef]
  474. Xue, J.; Xiao, T.; Wei, S.; Sun, J.; Zou, Z.; Shi, M.; Sun, Q.; Dai, X.; Wu, L.; Li, J.; et al. miR-21-regulated M2 polarization of macrophage is involved in arsenicosis-induced hepatic fibrosis through the activation of hepatic stellate cells. J. Cell. Physiol. 2021, 236, 6025–6041. [Google Scholar] [CrossRef]
  475. Shen, D.; He, Z. Mesenchymal stem cell-derived exosomes regulate the polarization and inflammatory response of macrophages via miR-21-5p to promote repair after myocardial reperfusion injury. Ann. Transl. Med. 2021, 9, 1323. [Google Scholar] [CrossRef]
  476. Li, J.; Wei, C.; Yang, Y.; Gao, Z.; Guo, Z.; Qi, F. Apoptotic bodies extracted from adipose mesenchymal stem cells carry microRNA-21-5p to induce M2 polarization of macrophages and augment skin wound healing by targeting KLF6. Burns 2022, 48, 1893–1908. [Google Scholar] [CrossRef]
  477. Xi, J.; Huang, Q.; Wang, L.; Ma, X.; Deng, Q.; Kumar, M.; Zhou, Z.; Li, L.; Zeng, Z.; Young, K.H.; et al. miR-21 depletion in macrophages promotes tumoricidal polarization and enhances PD-1 immunotherapy. Oncogene 2018, 37, 3151–3165. [Google Scholar] [CrossRef]
  478. Lu, J.; Xie, L.; Sun, S. The inhibitor miR-21 regulates macrophage polarization in an experimental model of chronic obstructive pulmonary disease. Tob. Induc. Dis. 2021, 19, 69. [Google Scholar] [CrossRef]
  479. Lee, H.Y.; Hur, J.; Kang, J.Y.; Rhee, C.K.; Lee, S.Y. MicroRNA-21 inhibition suppresses alveolar M2 macrophages in an ovalbumin-induced allergic asthma mice model. Allergy Asthma Immunol. Res. 2021, 13, 312–329. [Google Scholar] [CrossRef]
  480. Chen, Q.; Li, Y.; Gao, W.; Chen, L.; Xu, W.; Zhu, X. Exosome-mediated crosstalk between tumor and tumor-associated macrophages. Front. Mol. Biosci. 2021, 8, 764222. [Google Scholar] [CrossRef]
  481. Liu, W.; Zhu, M.; Wang, H.; Wang, W.; Lu, Y. Diffuse large B cell lymphoma-derived extracellular vesicles educate macrophages to promote tumours progression by increasing PGC-1β. Scand. J. Immunol. 2020, 91, e12841. [Google Scholar] [CrossRef]
  482. Ling, H.Y.; Yang, Z.; Wang, P.J.; Sun, Y.; Ju, S.G.; Li, J.; Fu, J.X. Diffuse large B-cell lymphoma-derived exosomes push macrophage polarization toward M2 phenotype via GP130/STAT3 signaling pathway. Chem. Biol. Interact. 2022, 352, 109779. [Google Scholar] [CrossRef]
  483. Ren, W.; Hou, J.; Yang, C.; Wang, H.; Wu, S.; Wu, Y.; Zhao, X.; Lu, C. Extracellular vesicles secreted by hypoxia pre-challenged mesenchymal stem cells promote non-small cell lung cancer cell growth and mobility as well as macrophage M2 polarization via miR-21-5p delivery. J. Exp. Clin. Cancer Res. 2019, 38, 62. [Google Scholar] [CrossRef] [Green Version]
  484. Hsieh, C.H.; Tai, S.K.; Yang, M.H. Snail-overexpressing cancer cells promote M2-like polarization of tumor-associated macrophages by delivering miR-21-abundant exosomes. Neoplasia 2018, 20, 775–788. [Google Scholar] [CrossRef] [PubMed]
  485. Lin, F.; Yin, H.B.; Li, X.Y.; Zhu, G.M.; He, W.Y.; Gou, X. Bladder cancer cell-secreted exosomal miR-21 activates the PI3K/AKT pathway in macrophages to promote cancer progression. Int. J. Oncol. 2020, 56, 151–164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  486. Yao, M.; Cui, B.; Zhang, W.; Ma, W.; Zhao, G.; Xing, L. Exosomal miR-21 secreted by IL-1β-primed-mesenchymal stem cells induces macrophage M2 polarization and ameliorates sepsis. Life Sci. 2021, 264, 118658. [Google Scholar] [CrossRef] [PubMed]
  487. Jin, J.; Yu, G. Hypoxic lung cancer cell-derived exosomal miR-21 mediates macrophage M2 polarization and promotes cancer cell proliferation through targeting IRF1. World J. Surg. Oncol. 2022, 20, 241. [Google Scholar] [CrossRef] [PubMed]
  488. Iliopoulos, D.; Jaeger, S.A.; Hirsch, H.A.; Bulyk, M.L.; Struhl, K. STAT3 activation of miR-21 and miR-181b-1 via PTEN and CYLD are part of the epigenetic switch linking inflammation to cancer. Mol. Cell. 2010, 39, 493–506. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  489. Kumarswamy, R.; Volkmann, I.; Thum, T. Regulation and function of miRNA-21 in health and disease. RNA Biol. 2011, 8, 706–713. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  490. van der Fits, L.; van Kester, M.S.; Qin, Y.; Out-Luiting, J.J.; Smit, F.; Zoutman, W.H.; Willemze, R.; Tensen, C.P.; Vermeer, M.H. MicroRNA-21 expression in CD4+ T cells is regulated by STAT3 and is pathologically involved in Sézary syndrome. J. Investig. Dermatol. 2011, 131, 762–768. [Google Scholar] [CrossRef] [Green Version]
  491. Chuang, H.Y.; Su, Y.K.; Liu, H.W.; Chen, C.H.; Chiu, S.C.; Cho, D.Y.; Lin, S.Z.; Chen, Y.S.; Lin, C.M. Preclinical evidence of STAT3 inhibitor pacritinib overcoming temozolomide resistance via downregulating miR-21-enriched exosomes from M2 glioblastoma-associated macrophages. J. Clin. Med. 2019, 8, 959. [Google Scholar] [CrossRef] [Green Version]
  492. Lan, J.; Sun, L.; Xu, F.; Liu, L.; Hu, F.; Song, D.; Hou, Z.; Wu, W.; Luo, X.; Wang, J.; et al. M2 macrophage-derived exosomes promote cell migration and invasion in colon cancer. Cancer Res. 2019, 79, 146–158. [Google Scholar] [CrossRef] [Green Version]
  493. Pu, J.; Xu, Z.; Nian, J.; Fang, Q.; Yang, M.; Huang, Y.; Li, W.; Ge, B.; Wang, J.; Wei, H. M2 macrophage-derived extracellular vesicles facilitate CD8+ T cell exhaustion in hepatocellular carcinoma via the miR-21-5p/YOD1/YAP/β-catenin pathway. Cell Death Discov. 2021, 7, 182. [Google Scholar] [CrossRef]
  494. Chang, J.; Li, H.; Zhu, Z.; Mei, P.; Hu, W.; Xiong, X.; Tao, J. microRNA-21-5p from M2 macrophage-derived extracellular vesicles promotes the differentiation and activity of pancreatic cancer stem cells by mediating KLF3. Cell Biol. Toxicol. 2022, 38, 577–590. [Google Scholar] [CrossRef]
  495. Howard, K.M.; Jati Kusuma, R.; Baier, S.R.; Friemel, T.; Markham, L.; Vanamala, J.; Zempleni, J. Loss of miRNAs during processing and storage of cow’s (Bos taurus) milk. J. Agric. Food Chem. 2015, 63, 588–592. [Google Scholar] [CrossRef] [Green Version]
  496. Li, Z.; Hassan, M.Q.; Jafferji, M.; Aqeilan, R.I.; Garzon, R.; Croce, C.M.; van Wijnen, A.J.; Stein, J.L.; Stein, G.S.; Lian, J.B. Biological functions of miR-29b contribute to positive regulation of osteoblast differentiation. J. Biol. Chem. 2009, 284, 15676–15684. [Google Scholar] [CrossRef] [Green Version]
  497. Griffiths-Jones, S. miRBase: MicroRNA sequences and annotation. Curr. Protoc. Bioinform. 2010, 29, 12.9.1–12.9.10. [Google Scholar] [CrossRef]
  498. Zhang, P.P.; Wang, Y.C.; Cheng, C.; Zhang, F.; Ding, D.Z.; Chen, D.K. Runt-related transcription factor 2 influences cell adhesion-mediated drug resistance and cell proliferation in B-cell non-Hodgkin’s lymphoma and multiple myeloma. Leuk. Res. 2020, 92, 106340. [Google Scholar] [CrossRef]
  499. Hines, M.J.; Coffre, M.; Mudianto, T.; Panduro, M.; Wigton, E.J.; Tegla, C.; Osorio-Vasquez, V.; Kageyama, R.; Benhamou, D.; Perez, O.; et al. miR-29 sustains B cell survival and controls terminal differentiation via regulation of PI3K signaling. Cell Rep. 2020, 33, 108436. [Google Scholar] [CrossRef]
  500. Anastasiadou, E.; Boccellato, F.; Vincenti, S.; Rosato, P.; Bozzoni, I.; Frati, L.; Faggioni, A.; Presutti, C.; Trivedi, P. Epstein-Barr virus encoded LMP1 downregulates TCL1 oncogene through miR-29b. Oncogene 2010, 29, 1316–1328. [Google Scholar] [CrossRef] [Green Version]
  501. Teitell, M.; Damore, M.A.; Sulur, G.G.; Turner, D.E.; Stern, M.H.; Said, J.W.; Denny, C.T.; Wall, R. TCL1 oncogene expression in AIDS-related lymphomas and lymphoid tissues. Proc. Natl. Acad. Sci. USA 1999, 96, 9809–9814. [Google Scholar] [CrossRef] [Green Version]
  502. Said, J.W.; Hoyer, K.K.; French, S.W.; Rosenfelt, L.; Garcia-Lloret, M.; Koh, P.J.; Cheng, T.C.; Sulur, G.G.; Pinkus, G.S.; Kuehl, W.M.; et al. TCL1 oncogene expression in B cell subsets from lymphoid hyperplasia and distinct classes of B cell lymphoma. Lab. Investig. 2001, 81, 555–564. [Google Scholar] [CrossRef] [Green Version]
  503. Izumi, H.; Kosaka, N.; Shimizu, T.; Sekine, K.; Ochiya, T.; Takase, M. Bovine milk contains microRNA and messenger RNA that are stable under degradative conditions. J. Dairy Sci. 2012, 95, 4831–4841. [Google Scholar] [CrossRef] [Green Version]
  504. Wang, L.; Shi, Z.; Wang, X.; Mu, S.; Xu, X.; Shen, L.; Li, P. Protective effects of bovine milk exosomes against oxidative stress in IEC-6 cells. Eur. J. Nutr. 2021, 60, 317–327. [Google Scholar] [CrossRef] [PubMed]
  505. Hatton, O.; Smith, M.M.; Alexander, M.; Mandell, M.; Sherman, C.; Stesney, M.W.; Hui, S.T.; Dohrn, G.; Medrano, J.; Ringwalt, K.; et al. Epstein-Barr virus latent membrane protein 1 regulates host B cell microRNA-155 and its target FOXO3a via PI3K p110α activation. Front. Microbiol. 2019, 10, 2692. [Google Scholar] [CrossRef] [PubMed]
  506. Kapoor, I.; Li, Y.; Sharma, A.; Zhu, H.; Bodo, J.; Xu, W.; His, E.D.; Hill, B.T.; Almasan, A. Resistance to BTK inhibition by ibrutinib can be overcome by preventing FOXO3a nuclear export and PI3K/AKT activation in B-cell lymphoid malignancies. Cell Death Dis. 2019, 10, 924. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  507. Campos, A.; Sharma, S.; Obermair, A.; Salomon, C. Extracellular vesicle-associated miRNAs and chemoresistance: A systematic review. Cancers 2021, 13, 4608. [Google Scholar] [CrossRef] [PubMed]
  508. Liao, T.L.; Hsieh, S.L.; Chen, Y.M.; Chen, H.H.; Liu, H.J.; Lee, H.C.; Chen, D.Y. Rituximab may cause increased hepatitis C virus viremia in rheumatoid arthritis patients through declining exosomal microRNA-155. Arthritis Rheumatol. 2018, 70, 1209–1219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  509. Rainy, N.; Zayoud, M.; Kloog, Y.; Rechavi, O.; Goldstein, I. Viral oncomiR spreading between B and T cells is employed by Kaposi’s sarcoma herpesvirus to induce non-cell-autonomous target gene regulation. Oncotarget 2016, 7, 41870–41884. [Google Scholar] [CrossRef]
  510. Zheng, Z.; Sun, R.; Zhao, H.J.; Fu, D.; Zhong, H.J.; Weng, X.Q.; Qu, B.; Zhao, Y.; Wang, L.; Zhao, W.L. MiR155 sensitized B-lymphoma cells to anti-PD-L1 antibody via PD-1/PD-L1-mediated lymphoma cell interaction with CD8+T cells. Mol. Cancer. 2019, 18, 54. [Google Scholar] [CrossRef]
  511. Poles, W.A.; Nishi, E.E.; de Oliveira, M.B.; Eugênio, A.I.P.; de Andrade, T.A.; Campos, A.H.F.M.; de Campos, R.R.; Vassallo, J.; Alves, A.C.; Scapulatempo Neto, C.; et al. Targeting the polarization of tumor-associated macrophages and modulating mir-155 expression might be a new approach to treat diffuse large B-cell lymphoma of the elderly. Cancer Immunol. Immunother. 2019, 68, 269–282. [Google Scholar] [CrossRef]
  512. Tingö, L.; Ahlberg, E.; Johansson, L.; Pedersen, S.A.; Chawla, K.; Sætrom, P.; Cione, E.; Simpson, M.R. Non-coding RNAs in human breast milk: A systematic review. Front. Immunol. 2021, 12, 725323. [Google Scholar] [CrossRef]
  513. Kirchner, B.; Pfaffl, M.W.; Dumpler, J.; Von Mutius, E.; Ege, M.J. MicroRNA in native and processed cow’s milk and its implication for the farm milk effect on asthma. J. Allergy Clin. Immunol. 2016, 137, 1893–1895. [Google Scholar] [CrossRef] [Green Version]
  514. Chutipongtanate, S.; Morrow, A.L.; Newburg, D.S. Human milk extracellular vesicles: A biological system with clinical implications. Cells 2022, 11, 2345. [Google Scholar] [CrossRef]
  515. Benmoussa, A.; Ly, S.; Shan, S.T.; Laugier, J.; Boilard, E.; Gilbert, C.; Provost, P. A subset of extracellular vesicles carries the bulk of microRNAs in commercial dairy cow’s milk. J. Extracell. Vesicles 2017, 6, 1401897. [Google Scholar] [CrossRef] [Green Version]
  516. Zhou, Q.; Li, M.; Wang, X.; Li, Q.; Wang, T.; Zhu, Q.; Zhou, X.; Wang, X.; Gao, X.; Li, X. Immune-related microRNAs are abundant in breast milk exosomes. Int. J. Biol. Sci. 2012, 8, 118–123. [Google Scholar] [CrossRef]
  517. Guo, M.M.; Zhang, K.; Zhang, J.H. Human breast milk-derived exosomal miR-148a-3p protects against necrotizing enterocolitis by regulating p53 and sirtuin 1. Inflammation 2022, 45, 1254–1268. [Google Scholar] [CrossRef]
  518. Gu, Y.; Li, M.; Wang, T.; Liang, Y.; Zhong, Z.; Wang, X.; Zhou, Q.; Chen, L.; Lang, Q.; He, Z.; et al. Lactation-related microRNA expression profiles of porcine breast milk exosomes. PLoS ONE 2012, 7, e43691. [Google Scholar] [CrossRef]
  519. Melnik, B.C.; Schmitz, G. MicroRNAs: Milk’s epigenetic regulators. Best. Pract. Res. Clin. Endocrinol. Metab. 2017, 31, 427–442. [Google Scholar] [CrossRef]
  520. Melnik, B.C. Milk disrupts p53 and DNMT1, the guardians of the genome: Implications for acne vulgaris and prostate cancer. Nutr. Metab. 2017, 14, 55. [Google Scholar] [CrossRef] [Green Version]
  521. Pappas, K.; Xu, J.; Zairis, S.; Resnick-Silverman, L.; Abate, F.; Steinbach, N.; Ozturk, S.; Saal, L.H.; Su, T.; Cheung, P.; et al. p53 maintains baseline expression of multiple tumor suppressor genes. Mol. Cancer Res. 2017, 15, 1051–1062. [Google Scholar] [CrossRef] [Green Version]
  522. Izquierdo, E.; Vorholt, D.; Blakemore, S.; Sackey, B.; Nolte, J.L.; Barbarino, V.; Schmitz, J.; Nickel, N.; Bachurski, D.; Lobastova, L.; et al. Extracellular vesicles and PD-L1 suppress macrophages, inducing therapy resistance in TP53-deficient B-cell malignancies. Blood 2022, 139, 3617–3629. [Google Scholar] [CrossRef]
  523. Qingjuan, L.; Xiaojuan, F.; Wei, Z.; Chao, W.; Pengpeng, K.; Hongbo, L.; Sanbing, Z.; Jun, H.; Min, Y.; Shuxia, L. miR-148a-3p overexpression contributes to glomerular cell proliferation by targeting PTEN in lupus nephritis. Am. J. Physiol. Cell. Physiol. 2016, 310, C470–C478. [Google Scholar] [CrossRef] [Green Version]
  524. Huang, F.; Zhao, J.L.; Wang, L.; Gao, C.C.; Liang, S.Q.; An, D.J.; Bai, J.; Chen, Y.; Han, H.; Qin, H.Y. miR-148a-3p mediates notch signaling to promote the differentiation and M1 activation of macrophages. Front. Immunol. 2017, 8, 1327. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  525. He, H.; Cai, M.; Zhu, J.; Xiao, W.; Liu, B.; Shi, Y.; Yang, X.; Liang, X.; Zheng, T.; Hu, S.; et al. miR-148a-3p promotes rabbit preadipocyte differentiation by targeting PTEN. In Vitro Cell Dev. Biol. Anim. 2018, 54, 241–249. [Google Scholar] [CrossRef] [PubMed]
  526. Reif, S.; Elbaum Shiff, Y.; Golan-Gerstl, R. Milk-derived exosomes (MDEs) have a different biological effect on normal fetal colon epithelial cells compared to colon tumor cells in a miRNA-dependent manner. J. Transl. Med. 2019, 17, 325. [Google Scholar] [CrossRef] [PubMed]
  527. Ma, Y.; Zhang, P.; Gao, Y.; Fan, H.; Zhang, M.; Wu, J. Evaluation of AKT phosphorylation and PTEN loss and their correlation with the resistance of rituximab in DLBCL. Int. J. Clin. Exp. Pathol. 2015, 8, 14875–14884. [Google Scholar]
  528. Cui, W.; Ma, M.; Zheng, S.; Ma, Z.; Su, L.; Zhang, W. PIK3CA amplification and PTEN loss in diffused large B-cell lymphoma. Oncotarget 2017, 8, 66237–66247. [Google Scholar] [CrossRef]
  529. Wang, X.; Cao, X.; Sun, R.; Tang, C.; Tzankov, A.; Zhang, J.; Manyam, G.C.; Xiao, M.; Miao, Y.; Jabbar, K.; et al. Clinical significance of PTEN deletion, mutation, and loss of PTEN expression in de novo diffuse large B-cell lymphoma. Neoplasia 2018, 20, 574–593. [Google Scholar] [CrossRef]
  530. Ben Younes, K.; Doghri, R.; Mrad, K.; Bedhiafi, W.; Benammar-Elgaaied, A.; Sola, B.; Ben Aissa-Fennira, F. PTEN loss and cyclin A2 upregulation define a PI3K/AKT pathway activation in Helicobacter pylori-induced MALT and DLBCL gastric lymphoma with features of MALT. Appl. Immunohistochem. Mol. Morphol. 2021, 29, 56–61. [Google Scholar] [CrossRef]
  531. Leeman-Neill, R.J.; Bhagat, G. BCL6 as a therapeutic target for lymphoma. Expert Opin. Ther. Targets 2018, 22, 143–152. [Google Scholar] [CrossRef]
  532. Xiao, F.; Rui, K.; Shi, X.; Wu, H.; Cai, X.; Lui, K.O.; Lu, Q.; Ballestar, E.; Tian, J.; Zou, H.; et al. Epigenetic regulation of B cells and its role in autoimmune pathogenesis. Cell. Mol. Immunol. 2022, 19, 1215–1234. [Google Scholar] [CrossRef]
  533. Wigton, E.J.; Ansel, K.M. Noncoding RNAs in B cell responses. RNA Biol. 2021, 18, 633–639. [Google Scholar] [CrossRef]
  534. Melnik, B.C.; Schmitz, G. Milk’s role as an epigenetic regulator in health and disease. Diseases 2017, 5, 12. [Google Scholar] [CrossRef]
  535. Teng, G.; Hakimpour, P.; Landgraf, P.; Rice, A.; Tuschl, T.; Casellas, R.; Papavasiliou, F.N. MicroRNA-155 is a negative regulator of activation-induced cytidine deaminase. Immunity 2008, 28, 621–629. [Google Scholar] [CrossRef] [Green Version]
  536. Borchert, G.M.; Holton, N.W.; Larson, E.D. Repression of human activation induced cytidine deaminase by miR-93 and miR-155. BMC Cancer 2011, 11, 347. [Google Scholar] [CrossRef] [Green Version]
  537. Frasca, D.; Diaz, A.; Romero, M.; Ferracci, F.; Blomberg, B.B. MicroRNAs miR-155 and miR-16 decrease AID and E47 in B Cells from elderly individuals. J. Immunol. 2015, 195, 2134–2140. [Google Scholar] [CrossRef] [Green Version]
  538. Recaldin, T.; Hobson, P.S.; Mann, E.H.; Ramadani, F.; Cousins, D.J.; Lavender, P.; Fear, D.J. miR-29b directly targets activation-induced cytidine deaminase in human B cells and can limit its inappropriate expression in naïve B cells. Mol. Immunol. 2018, 101, 419–428. [Google Scholar] [CrossRef] [Green Version]
  539. Alles, J.; Ludwig, N.; Rheinheimer, S.; Leidinger, P.; Grässer, F.A.; Keller, A.; Meese, E. MiR-148a impairs Ras/ERK signaling in B lymphocytes by targeting SOS proteins. Oncotarget 2017, 8, 56417–56427. [Google Scholar] [CrossRef] [Green Version]
  540. Yasuda, T.; Kometani, K.; Takahashi, N.; Imai, Y.; Aiba, Y.; Kurosaki, T. ERKs induce expression of the transcriptional repressor Blimp-1 and subsequent plasma cell differentiation. Sci. Signal. 2011, 4, ra25. [Google Scholar] [CrossRef]
  541. Allman, D.M.; Cancro, M.P. pERKing up the BLIMP in plasma cell differentiation. Sci. Signal. 2011, 4, pe21. [Google Scholar] [CrossRef]
  542. Minnich, M.; Tagoh, H.; Bönelt, P.; Axelsson, E.; Fischer, M.; Cebolla, B.; Tarakhovsky, A.; Nutt, S.L.; Jaritz, M.; Busslinger, M. Multifunctional role of the transcription factor Blimp-1 in coordinating plasma cell differentiation. Nat. Immunol. 2016, 17, 331–343. [Google Scholar] [CrossRef] [Green Version]
  543. Goodman, A.; Patel, S.P.; Kurzrock, R. PD-1-PD-L1 immune-checkpoint blockade in B-cell lymphomas. Nat. Rev. Clin. Oncol. 2017, 14, 203–220. [Google Scholar] [CrossRef]
  544. Vari, F.; Arpon, D.; Keane, C.; Hertzberg, M.S.; Talaulikar, D.; Jain, S.; Cui, Q.; Han, E.; Tobin, J.; Bird, R.; et al. Immune evasion via PD-1/PD-L1 on NK cells and monocyte/macrophages is more prominent in Hodgkin lymphoma than DLBCL. Blood 2018, 131, 1809–1819. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  545. Wang, Y.; Wu, L.; Tian, C.; Zhang, Y. PD-1-PD-L1 immune-checkpoint blockade in malignant lymphomas. Ann. Hematol. 2018, 97, 229–237. [Google Scholar] [CrossRef] [PubMed]
  546. Xu-Monette, Z.Y.; Xiao, M.; Au, Q.; Padmanabhan, R.; Xu, B.; Hoe, N.; Rodríguez-Perales, S.; Torres-Ruiz, R.; Manyam, G.C.; Visco, C.; et al. Immune profiling and quantitative analysis decipher the clinical role of immune-checkpoint expression in the tumor immune microenvironment of DLBCL. Cancer Immunol. Res. 2019, 7, 644–657. [Google Scholar] [CrossRef] [PubMed]
  547. McCord, R.; Bolen, C.R.; Koeppen, H.; Kadel, E.E.; Oestergaard, M.Z.; Nielsen, T.; Sehn, L.H.; Venstrom, J.M. PD-L1 and tumor-associated macrophages in de novo DLBCL. Blood Adv. 2019, 3, 531–540. [Google Scholar] [CrossRef] [Green Version]
  548. Song, M.K.; Park, B.B.; Uhm, J. Understanding immune evasion and therapeutic targeting associated with PD-1/PD-L1 pathway in diffuse large B-cell lymphoma. Int. J. Mol. Sci. 2019, 20, 1326. [Google Scholar] [CrossRef] [Green Version]
  549. Xie, W.; Medeiros, L.J.; Li, S.; Yin, C.C.; Khoury, J.D.; Xu, J. PD-1/PD-L1 pathway and its blockade in patients with classic Hodgkin lymphoma and Non-Hodgkin large-cell lymphomas. Curr. Hematol. Malig. Rep. 2020, 15, 372–381. [Google Scholar] [CrossRef]
  550. Ashizawa, M.; Okayama, H.; Ishigame, T.; Thar Min, A.K.; Saito, K.; Ujiie, D.; Murakami, Y.; Kikuchi, T.; Nakayama, Y.; Noda, M.; et al. miRNA-148a-3p regulates immunosuppression in DNA mismatch repair-deficient colorectal cancer by targeting PD-L1. Mol. Cancer Res. 2019, 17, 1403–1413. [Google Scholar] [CrossRef] [Green Version]
  551. Wang, Q.; Lin, W.; Tang, X.; Li, S.; Guo, L.; Lin, Y.; Kwok, H.F. The roles of microRNAs in regulating the expression of PD-1/PD-L1 immune checkpoint. Int. J. Mol. Sci. 2017, 18, 2540. [Google Scholar] [CrossRef] [Green Version]
  552. Huang, C.; Melnick, A. Mechanisms of action of BCL6 during germinal center B cell development. Sci. China Life Sci. 2015, 58, 1226–1232. [Google Scholar] [CrossRef] [Green Version]
  553. Ye, B.H.; Lista, F.; Lo Coco, F.; Knowles, D.M.; Offit, K.; Chaganti, R.S.; Dalla-Favera, R. Alterations of a zinc finger-encoding gene, BCL-6, in diffuse large-cell lymphoma. Science 1993, 262, 747–750. [Google Scholar] [CrossRef]
  554. Basso, K.; Dalla-Favera, R. BCL6: Master regulator of the germinal center reaction and key oncogene in B cell lymphomagenesis. Adv. Immunol. 2010, 105, 193–210. [Google Scholar]
  555. van Esch, B.C.A.M.; Porbahaie, M.; Abbring, S.; Garssen, J.; Potaczek, D.P.; Savelkoul, H.F.J.; van Neerven, R.J.J. The impact of milk and its components on epigenetic programming of immune function in early life and beyond: Implications for allergy and asthma. Front. Immunol. 2020, 11, 2141. [Google Scholar] [CrossRef]
  556. Duy, C.; Yu, J.J.; Nahar, R.; Swaminathan, S.; Kweon, S.M.; Polo, J.M.; Valls, E.; Klemm, L.; Shojaee, S.; Cerchietti, L.; et al. BCL6 is critical for the development of a diverse primary B cell repertoire. J. Exp. Med. 2010, 207, 1209–1221. [Google Scholar] [CrossRef] [Green Version]
  557. Crotty, S.; Johnston, R.J.; Schoenberger, S.P. Effectors and memories: Bcl-6 and Blimp-1 in T and B lymphocyte differentiation. Nat. Immunol. 2010, 11, 114–120. [Google Scholar] [CrossRef] [Green Version]
  558. Atyeo, C.; Alter, G. The multifaceted roles of breast milk antibodies. Cell 2021, 184, 1486–1499. [Google Scholar] [CrossRef]
  559. Yu, J.C.; Khodadadi, H.; Malik, A.; Davidson, B.; Salles, É.D.S.L.; Bhatia, J.; Hale, V.L.; Baban, B. Innate immunity of neonates and infants. Front. Immunol. 2018, 9, 1759. [Google Scholar] [CrossRef] [Green Version]
  560. Yu, H.R.; Huang, L.H.; Li, S.C. Roles of microRNA in the immature immune system of neonates. Cancer Lett. 2018, 433, 99–106. [Google Scholar] [CrossRef]
  561. Pettersson, S.; Pobor, G.; Coutinho, A. Ontogenic development of B cell reactivities to cooperative cell signals: Dissociation between proliferation and antibody secretion. Eur. J. Immunol. 1982, 12, 653–658. [Google Scholar] [CrossRef]
  562. Melnik, B.C. Milk exosomal miRNAs: Potential drivers of AMPK-to-mTORC1 switching in β-cell de-differentiation of type 2 diabetes mellitus. Nutr. Metab. 2019, 16, 85. [Google Scholar] [CrossRef]
  563. Melnik, B.C.; Schmitz, G. Milk exosomal microRNAs: Postnatal promoters of β cell proliferation but potential inducers of β cell de-differentiation in adult life. Int. J. Mol. Sci. 2022, 23, 11503. [Google Scholar] [CrossRef]
  564. Dominguez, P.M.; Teater, M.; Chambwe, N.; Kormaksson, M.; Redmond, D.; Ishii, J.; Vuong, B.; Chaudhuri, J.; Melnick, A.; Vasanthakumar, A.; et al. DNA methylation dynamics of germinal center B cells are mediated by AID. Cell Rep. 2015, 12, 2086–2098. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  565. Teater, M.; Dominguez, P.M.; Redmond, D.; Chen, Z.; Ennishi, D.; Scott, D.W.; Cimmino, L.; Ghione, P.; Chaudhuri, J.; Gascoyne, R.D.; et al. AICDA drives epigenetic heterogeneity and accelerates germinal center-derived lymphomagenesis. Nat. Commun. 2018, 9, 222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  566. You, H.; Xu-Monette, Z.Y.; Wei, L.; Nunns, H.; Nagy, M.L.; Bhagat, G.; Fang, X.; Zhu, F.; Visco, C.; Tzankov, A.; et al. Genomic complexity is associated with epigenetic regulator mutations and poor prognosis in diffuse large B-cell lymphoma. Oncoimmunology 2021, 10, 1928365. [Google Scholar] [CrossRef] [PubMed]
  567. Çakan, E.; Gunaydin, G. Activation induced cytidine deaminase: An old friend with new faces. Front. Immunol. 2022, 13, 965312. [Google Scholar] [CrossRef]
  568. Kozloski, G.A.; Lossos, I.S. LymphomiRs: MicroRNAs with regulatory roles in lymphomas. Curr. Opin. Hematol. 2015, 22, 362–368. [Google Scholar] [CrossRef]
  569. Aune, D.; Lau, R.; Chan, D.S.M.; Vieira, R.; Greenwood, D.C.; Kampman, E.; Norat, T. Dairy products and colorectal cancer risk: A systematic review and meta-analysis of cohort studies. Ann. Oncol. 2012, 23, 37–45. [Google Scholar] [CrossRef]
  570. Bakken, T.; Braaten, T.; Olsen, A.; Hjartåker, A.; Lund, E.; Skeie, G. Milk and risk of colorectal, colon and rectal cancer in the Norwegian Women and Cancer (NOWAC) Cohort Study. Br. J. Nutr. 2018, 119, 1274–1285. [Google Scholar] [CrossRef] [Green Version]
  571. Barrubés, L.; Babio, N.; Becerra-Tomás, N.; Rosique-Esteban, N.; Salas-Salvadó, J. Association between dairy product consumption and colorectal cancer risk in adults: A systematic review and meta-analysis of epidemiologic studies. Adv. Nutr. 2019, 10, S190–S211, Erratum in: Adv. Nutr. 2020, 11, 1055–1057. [Google Scholar] [CrossRef]
  572. Lumsden, A.L.; Mulugeta, A.; Hyppönen, E. Milk consumption and risk of twelve cancers: A large-scale observational and Mendelian randomisation study. Clin. Nutr. 2023, 42, 1–8. [Google Scholar] [CrossRef]
  573. Stremmel, W.; Weiskirchen, R.; Melnik, B.C. Milk exosomes prevent intestinal inflammation in a genetic mouse model of ulcerative colitis: A pilot experiment. Inflamm. Intest. Dis. 2020, 5, 117–123. [Google Scholar] [CrossRef]
  574. Reif, S.; Elbaum-Shiff, Y.; Koroukhov, N.; Shilo, I.; Musseri, M.; Golan-Gerstl, R. Cow and human milk-derived exosomes ameliorate colitis in DSS murine model. Nutrients 2020, 12, 2589. [Google Scholar] [CrossRef]
  575. Zhao, W.; Zheng, J.; Wei, G.; Yang, K.; Wang, G.; Sun, X. miR-148a inhibits cell proliferation and migration through targeting ErbB3 in colorectal cancer. Oncol. Lett. 2019, 18, 2530–2536. [Google Scholar] [CrossRef] [Green Version]
  576. Martino, E.; Balestrieri, A.; Mele, L.; Sardu, C.; Marfella, R.; D’Onofrio, N.; Campanile, G.; Balestrieri, M.L. Milk exosomal miR-27b worsen endoplasmic reticulum stress mediated colorectal cancer cell death. Nutrients 2022, 14, 5081. [Google Scholar] [CrossRef]
  577. Peng, X.; Chen, G.; Lv, B.; Lv, J. MicroRNA-148a/152 cluster restrains tumor stem cell phenotype of colon cancer via modulating CCT6A. Anticancer Drugs 2022, 33, e610–e621. [Google Scholar] [CrossRef]
  578. Melnik, B.C.; Stremmel, W.; Weiskirchen, R.; John, S.M.; Schmitz, G. Exosome-derived microRNAs of human milk and their effects on infant health and development. Biomolecules 2021, 11, 851. [Google Scholar] [CrossRef]
  579. Leroux, C.; Chervet, M.L.; German, J.B. Perspective: Milk microRNAs as important players in infant physiology and development. Adv. Nutr. 2021, 12, 1625–1635. [Google Scholar] [CrossRef]
  580. Melnik, B.C.; Schmitz, G. Exosomes of pasteurized milk: Potential pathogens of Western diseases. J. Transl. Med. 2019, 17, 3. [Google Scholar] [CrossRef] [Green Version]
  581. Melnik, B.C.; Weiskirchen, R.; Schmitz, G. Milk exosomal microRNAs: Friend or foe? A narrative review. ExRNA 2022, 4, 22. [Google Scholar] [CrossRef]
  582. Ghosh, S.; Iacucci, M. Diverse immune effects of bovine colostrum and benefits in human health and disease. Nutrients 2021, 13, 3798. [Google Scholar] [CrossRef]
  583. García-Martínez, J.; Pérez-Castillo, Í.M.; Salto, R.; López-Pedrosa, J.M.; Rueda, R.; Girón, M.D. Beneficial effects of bovine milk exosomes in metabolic interorgan cross-talk. Nutrients 2022, 14, 1442. [Google Scholar] [CrossRef]
  584. Cleveland Clinic. Health Essentials. A Healthy Diet for Diffuse Large B-Cell Lymphoma—Certain Foods Can Boost Your Well-Being During Treatment. 27 May 2022. Available online: https://health.clevelandclinic.org/diffuse-large-b-cell-lymphoma-diet/ (accessed on 15 March 2023).
  585. Christian, P.; Smith, E.R.; Lee, S.E.; Vargas, A.J.; Bremer, A.A.; Raiten, D.J. The need to study human milk as a biological system. Am. J. Clin. Nutr. 2021, 113, 1063–1072. [Google Scholar] [CrossRef] [PubMed]
  586. Wehbe, Z.; Kreydiyyeh, S. Cow’s milk may be delivering potentially harmful undetected cargoes to humans. Is it time to reconsider dairy recommendations? Nutr. Rev. 2022, 80, 874–888. [Google Scholar] [CrossRef] [PubMed]
  587. Klapper, W.; Kreuz, M.; Kohler, C.W.; Burkhardt, B.; Szczepanowski, M.; Salaverria, I.; Hummel, M.; Loeffler, M.; Pellissery, S.; Woessmann, W.; et al. Molecular mechanisms in Malignant Lymphomas Network Project of the Deutsche Krebshilfe. Patient age at diagnosis is associated with the molecular characteristics of diffuse large B-cell lymphoma. Blood 2012, 119, 1882–1887. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  588. American Cancer Society. Non-Hodgkin Lymphoma Risk Factors. Available online: https://www.cancer.org/cancer/non-hodgkin-lymphoma/causes-risks-prevention/risk-factors.html (accessed on 5 March 2023).
  589. Epperla, N.; Vaughn, J.L.; Othus, M.; Hallack, A.; Costa, L.J. Recent survival trends in diffuse large B-cell lymphoma--Have we made any progress beyond rituximab? Cancer Med. 2020, 9, 5519–5525. [Google Scholar] [CrossRef]
  590. Shi, Y.; Han, Y.; Yang, J.; Liu, P.; He, X.; Zhang, C.; Zhou, S.; Zhou, L.; Qin, Y.; Song, Y.; et al. Clinical features and outcomes of diffuse large B-cell lymphoma based on nodal or extranodal primary sites of origin: Analysis of 1085 WHO classified cases in a single institution in China. Chin. J. Cancer Res. 2019, 31, 152–161. [Google Scholar] [CrossRef]
  591. Torfadottir, J.E.; Steingrimsdottir, L.; Mucci, L.; Aspelund, T.; Kasperzyk, J.L.; Olafsson, O.; Fall, K.; Tryggvadottir, L.; Harris, T.B.; Launer, L.; et al. Milk intake in early life and risk of advanced prostate cancer. Am. J. Epidemiol. 2012, 175, 144–153. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Potential synergism of milk-induced phosphatidylinositol-3-kinase (PI3K)-AKT-mechanistic target of rapamycin complex 1 (mTORC1) signaling and overstimulated PI3K-AKT-mTORC1 signaling in diffuse large B-cell lymphoma (DLBCL). B-cell receptor (BCR) is commonly activated in DLBCL and stimulates the mTORC1 pathway. Milk-derived peptides are known agents interacting with BCR. Insulin-like growth factor 1 (IGF-1)/IGF-1 receptor (IGF1R) signaling is also activated in DLBCL. Milk contains IGF-1 and stimulates IGF-1 synthesis in human milk consumers. IGF-1/IGF1R signaling activates mTORC1. Expression or function of phosphatase and tensin homolog (PTEN) is commonly suppressed in DLBCL and is also targeted by microRNAs (miRs), especially miR-21. DLBCL cells frequently overexpress L-type amino acid transporter 1 (LAT1), which plays a crucial role in intracellular uptake of leucine (Leu), which via Ragulator (RAG) activates mTORC1. Leu is an abundant amino acid of milk proteins. Glutamine (Gln) is also enriched in milk proteins. The glutaminolysis pathway is activated in DLBCL cells. The end product α-ketoglutarate (αKG) via RAG as well activates mTORC1, which finally promotes the translation of the oncogene C-MYC, the key driver of cell proliferation. The activated kinase AKT stimulates YES1-associated transcriptional regulator (YES) of the HIPPO pathway found to be activated in DLBCL. Estrogen receptor β (ERβ) was also found to be overexpressed in subtypes DLBCLs. ERβ induces the expression of IGF1R and miR-21-5p. Milk-derived estrogens and milk contamination with bisphenol A (BPA) may contribute to ERβ signaling in DLBCL. AKT-mediated activation of mouse double minute 2 (MDM2) promotes the proteasomal degradation of p53, the guardian of the genome. p53 regulates the transcription of cyclin-dependent kinase inhibitor 1A (CDKN1A; p21) and of the anti-apoptotic protein survivin (BIRC5). TP53 is frequently downregulated in DLBCL. The oncogene B cell lymphoma 6 (BCL6) is commonly upregulated in DLBCL and functions as a negative regulator of both p53 and B lymphocyte-induced maturation protein 1 (BLIMP1, PRDM1). As such, milk-derived exosomal microRNAs play a key role in epigenetic regulation.
Figure 1. Potential synergism of milk-induced phosphatidylinositol-3-kinase (PI3K)-AKT-mechanistic target of rapamycin complex 1 (mTORC1) signaling and overstimulated PI3K-AKT-mTORC1 signaling in diffuse large B-cell lymphoma (DLBCL). B-cell receptor (BCR) is commonly activated in DLBCL and stimulates the mTORC1 pathway. Milk-derived peptides are known agents interacting with BCR. Insulin-like growth factor 1 (IGF-1)/IGF-1 receptor (IGF1R) signaling is also activated in DLBCL. Milk contains IGF-1 and stimulates IGF-1 synthesis in human milk consumers. IGF-1/IGF1R signaling activates mTORC1. Expression or function of phosphatase and tensin homolog (PTEN) is commonly suppressed in DLBCL and is also targeted by microRNAs (miRs), especially miR-21. DLBCL cells frequently overexpress L-type amino acid transporter 1 (LAT1), which plays a crucial role in intracellular uptake of leucine (Leu), which via Ragulator (RAG) activates mTORC1. Leu is an abundant amino acid of milk proteins. Glutamine (Gln) is also enriched in milk proteins. The glutaminolysis pathway is activated in DLBCL cells. The end product α-ketoglutarate (αKG) via RAG as well activates mTORC1, which finally promotes the translation of the oncogene C-MYC, the key driver of cell proliferation. The activated kinase AKT stimulates YES1-associated transcriptional regulator (YES) of the HIPPO pathway found to be activated in DLBCL. Estrogen receptor β (ERβ) was also found to be overexpressed in subtypes DLBCLs. ERβ induces the expression of IGF1R and miR-21-5p. Milk-derived estrogens and milk contamination with bisphenol A (BPA) may contribute to ERβ signaling in DLBCL. AKT-mediated activation of mouse double minute 2 (MDM2) promotes the proteasomal degradation of p53, the guardian of the genome. p53 regulates the transcription of cyclin-dependent kinase inhibitor 1A (CDKN1A; p21) and of the anti-apoptotic protein survivin (BIRC5). TP53 is frequently downregulated in DLBCL. The oncogene B cell lymphoma 6 (BCL6) is commonly upregulated in DLBCL and functions as a negative regulator of both p53 and B lymphocyte-induced maturation protein 1 (BLIMP1, PRDM1). As such, milk-derived exosomal microRNAs play a key role in epigenetic regulation.
Ijms 24 06102 g001
Figure 2. Illustrated mechanistic role of bovine milk-derived exosomes (MDE) and their microRNAs (miRs) in epigenetic signaling in diffuse large B-cell lymphoma (DLBCL). The key oncogene upregulated in most DLBCL is B cell lymphoma 6 (BCL6) activating the germinal center reaction and promoting B cell proliferation. The expression BCL6 is suppressed by a complex of DNA methyltransferase 1 (DNMT1) and activation-induced cytidine deaminase (encoded on AICDA), which catalyzes BCL6 promoter methylation. DNMT1 is a target of miR-148a, the most abundant miR of human and bovine milk and their MDEs. AICDA is targeted via miR-155 and miR-29b, abundant immune-regulatory miRs of human and bovine milk. MDE miRs may thus enhance BCL6 expression promoting B cell proliferation. BCL6 is a critical inhibitor of B lymphocyte-induced maturation protein 1 (BLIMP1, encoded on PRDM1), the key driver of B cell differentiation. miR-148a via suppression of SOS RAS/RAC guanine nucleotide exchange factor 1 and 2 (SOS1/2) inhibits ERK1/2-mediated expression of BLIMP1. Thus, miR-148a indirectly increases the ratio of BCL6/BLIMP1. BLIMP1 is directly suppressed via the let-7 family of miRs and miR-125b, further abundant miRs of human and bovine milk and their MDEs. Reduced expression of BLIMP1 enhances C-MYC expression, a key oncogene driving cell proliferation. C-MYC translation is dependent on the mechanistic target of rapamycin complex 1 (mTORC1). Negative regulators of mTORC1 are DEP domain-containing protein 6 (DEPTOR encoded on DEPDC6), phosphatase and tensin homolog (PTEN), and AMP-activated protein kinase (AMPK), which are targeted via abundant miR-155, miR-21 an miR-148a, respectively. In addition, miR-21 targets MAX dimerization protein 1 (MXD1), whereas miR-125b targets MAX dimerization protein 4 (MXD4), leading to sustained C-MYC activation. In addition, miR-148a and miR-125b target TP53, the key transcription factor of a multitude of tumor suppressor genes. miR-125b also targets interferon regulatory factor 4 (IRF4), a well-defined transcription factor obligatory required for the terminal differentiation of B cells to plasma cells. A further target of miR-125b is tumor necrosis factor alpha-induced protein 3 (TNFAIP3), also known as A20. TNFAIP3 is a negative regulator of nuclear factor kappa B (NF-κB) signaling, which is upregulated in most cases of DLBCL. Notably, miR-21 also targets TNFAIP3 as well as forkhead box O A1 (FOXO1A), programmed cell death 4 (PDCD4), and insulin-like growth factor-binding protein 3 (IGFBP3). Thus, the most abundant milk-derived miRs maintain mTORC1 activation, and promote the expression of BCL6 and NF-κB, but suppress cell cycle inhibitor p53 and transcription factors BLIMP1 and IRF4 involved in B cell differentiation and plasma cell and antibody formation. This signaling scenario may be useful for neonatal B cell proliferation during the period of lactation when high-affinity antibodies educated by the maternal immune system are provided to the infant via breastfeeding. However, persistent epigenetic milk miR signaling via continued exposure to bovine MDE-transferred miRs provided by the consumption of pasteurized cow’s milk may promote BCL6/BLIMP1-dependent lymphomagenesis.
Figure 2. Illustrated mechanistic role of bovine milk-derived exosomes (MDE) and their microRNAs (miRs) in epigenetic signaling in diffuse large B-cell lymphoma (DLBCL). The key oncogene upregulated in most DLBCL is B cell lymphoma 6 (BCL6) activating the germinal center reaction and promoting B cell proliferation. The expression BCL6 is suppressed by a complex of DNA methyltransferase 1 (DNMT1) and activation-induced cytidine deaminase (encoded on AICDA), which catalyzes BCL6 promoter methylation. DNMT1 is a target of miR-148a, the most abundant miR of human and bovine milk and their MDEs. AICDA is targeted via miR-155 and miR-29b, abundant immune-regulatory miRs of human and bovine milk. MDE miRs may thus enhance BCL6 expression promoting B cell proliferation. BCL6 is a critical inhibitor of B lymphocyte-induced maturation protein 1 (BLIMP1, encoded on PRDM1), the key driver of B cell differentiation. miR-148a via suppression of SOS RAS/RAC guanine nucleotide exchange factor 1 and 2 (SOS1/2) inhibits ERK1/2-mediated expression of BLIMP1. Thus, miR-148a indirectly increases the ratio of BCL6/BLIMP1. BLIMP1 is directly suppressed via the let-7 family of miRs and miR-125b, further abundant miRs of human and bovine milk and their MDEs. Reduced expression of BLIMP1 enhances C-MYC expression, a key oncogene driving cell proliferation. C-MYC translation is dependent on the mechanistic target of rapamycin complex 1 (mTORC1). Negative regulators of mTORC1 are DEP domain-containing protein 6 (DEPTOR encoded on DEPDC6), phosphatase and tensin homolog (PTEN), and AMP-activated protein kinase (AMPK), which are targeted via abundant miR-155, miR-21 an miR-148a, respectively. In addition, miR-21 targets MAX dimerization protein 1 (MXD1), whereas miR-125b targets MAX dimerization protein 4 (MXD4), leading to sustained C-MYC activation. In addition, miR-148a and miR-125b target TP53, the key transcription factor of a multitude of tumor suppressor genes. miR-125b also targets interferon regulatory factor 4 (IRF4), a well-defined transcription factor obligatory required for the terminal differentiation of B cells to plasma cells. A further target of miR-125b is tumor necrosis factor alpha-induced protein 3 (TNFAIP3), also known as A20. TNFAIP3 is a negative regulator of nuclear factor kappa B (NF-κB) signaling, which is upregulated in most cases of DLBCL. Notably, miR-21 also targets TNFAIP3 as well as forkhead box O A1 (FOXO1A), programmed cell death 4 (PDCD4), and insulin-like growth factor-binding protein 3 (IGFBP3). Thus, the most abundant milk-derived miRs maintain mTORC1 activation, and promote the expression of BCL6 and NF-κB, but suppress cell cycle inhibitor p53 and transcription factors BLIMP1 and IRF4 involved in B cell differentiation and plasma cell and antibody formation. This signaling scenario may be useful for neonatal B cell proliferation during the period of lactation when high-affinity antibodies educated by the maternal immune system are provided to the infant via breastfeeding. However, persistent epigenetic milk miR signaling via continued exposure to bovine MDE-transferred miRs provided by the consumption of pasteurized cow’s milk may promote BCL6/BLIMP1-dependent lymphomagenesis.
Ijms 24 06102 g002
Figure 3. Exosome crosstalk in the tumor microenvironment (TME) of diffuse large B-cell lymphoma (DLBCL). DLBCL cells secrete microRNA-21 (miR-21)-enriched exosomes into the TME to tumor-associated macrophages (TAM), which internalize these tumor-derived exosomes and promote macrophage M2 polarization, favoring tumor growth and progression. TAMs also secrete miR-21-enriched exosomes to DLBCL cells further promoting lymphomagenesis. Bovine milk-derived exosomes (MDEs) also transport miR-21 to recipient cells that may reach TAMs and/or DLBCL cells further augmenting miR-21-mediated oncogenic signaling.
Figure 3. Exosome crosstalk in the tumor microenvironment (TME) of diffuse large B-cell lymphoma (DLBCL). DLBCL cells secrete microRNA-21 (miR-21)-enriched exosomes into the TME to tumor-associated macrophages (TAM), which internalize these tumor-derived exosomes and promote macrophage M2 polarization, favoring tumor growth and progression. TAMs also secrete miR-21-enriched exosomes to DLBCL cells further promoting lymphomagenesis. Bovine milk-derived exosomes (MDEs) also transport miR-21 to recipient cells that may reach TAMs and/or DLBCL cells further augmenting miR-21-mediated oncogenic signaling.
Ijms 24 06102 g003
Figure 4. (A) Physiological interaction of human milk-derived exosomes (MDEs) and their microRNAs (miRs) in B lymphocyte proliferation via epigenetic upregulation of B cell lymphoma 6 (BCL6) and suppression of B lymphocyte-induced maturation protein 1 (BLIMP1), which suppresses B cell differentiation and antibody secretion. The neonatal immune system is immature and neonatal B cells are in a hyperproliferative undifferentiated state. The majority of specific antibodies are of maternal origin and are supplied via breastfeeding. Weaning, the physiological fading of MDE miRs, induces a key switch in milk-driven epigenetic regulation enhancing BLIMP1 expression and promoting antibody secretion by the infant’s matured immune system. (B) Persistent consumption of pasteurized cow’s milk maintains MDE miR-mediated epigenetic signaling promoting BCL6 expression and suppressing BLIMP1 expression, thus maintaining the hyperproliferative neonatal phenotype. Sustained proliferation is a hallmark of cancer, in this setting lymphomagenesis.
Figure 4. (A) Physiological interaction of human milk-derived exosomes (MDEs) and their microRNAs (miRs) in B lymphocyte proliferation via epigenetic upregulation of B cell lymphoma 6 (BCL6) and suppression of B lymphocyte-induced maturation protein 1 (BLIMP1), which suppresses B cell differentiation and antibody secretion. The neonatal immune system is immature and neonatal B cells are in a hyperproliferative undifferentiated state. The majority of specific antibodies are of maternal origin and are supplied via breastfeeding. Weaning, the physiological fading of MDE miRs, induces a key switch in milk-driven epigenetic regulation enhancing BLIMP1 expression and promoting antibody secretion by the infant’s matured immune system. (B) Persistent consumption of pasteurized cow’s milk maintains MDE miR-mediated epigenetic signaling promoting BCL6 expression and suppressing BLIMP1 expression, thus maintaining the hyperproliferative neonatal phenotype. Sustained proliferation is a hallmark of cancer, in this setting lymphomagenesis.
Ijms 24 06102 g004
Table 1. Recent epidemiological evidence for milk and dairy product consumption related to increased risk for B-cell lymphoma and DLBCL.
Table 1. Recent epidemiological evidence for milk and dairy product consumption related to increased risk for B-cell lymphoma and DLBCL.
Study CharacteristicsAssociated RiskReferences
Meta-analysis
14 case-control studies and two cohort studies
Milk consumption is related to increased risk of non-Hodgkin lymphoma (NHL) RR = 1.49 (95% CI: 1.08–2.06); risk of NHL increased by 5% and 6% for each 200 g/day increment in total dairy product and milk consumption, respectively[49]
An exposome-wide analysis based on the European Prospective Investigation into Cancer and Nutrition Study (n = 475,426)Positive association between dairy intake and risk of B-cell lymphoma and DLBCL[51]
Prospective China Kadoorie Biobank Study collecting ~0.5 million adults from 10 diverse (5 urban, 5 rural) areas across China during
2004–2008
Increased lymphoma risk related to dairy consumption (mainly milk) for urban HR = 1.09 (95% CI: 0.89–1.34) and rural regions HR = 1.45 (95% CI: 1.07–1.96)[50]
Table 2. Comparison of biologically active compounds related to milk consumption and DLBCL-associated pathology.
Table 2. Comparison of biologically active compounds related to milk consumption and DLBCL-associated pathology.
CompoundCow’s Milk IntakeReferencesDLBCLReferences
IGF-1Increased serum levels of
IGF-1 activate mTORC1
[67,68,69,70,71,72,73,74,75,76]Increased IGF-1/IGF1R
signaling
[61,62,63,64,65]
LeucineRich source of leucine
(10 g leucine/100 g milk protein) activates mTORC1
[92,93,94,95,96,97]High expression of leucine transporter LAT1 associated with poor prognosis[98,99]
GlutamineRich source of glutamine
9 g glutamine/100 g casein
[106,107]Glutaminolysis activates TCA cycle and mTORC1[101,102,103]
Milk-derived
peptides
B-cell epitopes stimulate BCR signaling[147,148,149,150,151,152,153,154,155]Persistent BCR activation augments mTORC1 signaling[108,109,110,111,112,113,128,129,130,131,132]
EstrogensIncreased levels in milk of persistently pregnant dairy cows[167,168,169,170,171,172,173,174]High ERβ expression is related to poor prognosis[157,158,159]
Bisphenol A (BPA)Contamination of commercial milk, weak estrogenic activity increases
IGF-1/mTORC1
[201,202,203,204,205]Lymphoma-promoting activities of BPA[37,182,183,184,185,186]
BMMFsViral single-stranded DNA[248,249]Suspected to promote cancer
induction early in life;
Role in DLBCL not yet experimentally investigated
[250,256]
Abbreviations: IGF-1, insulin-like growth factor 1; IGF1R, insulin-like growth factor 1 receptor; LAT1, L-type neutral amino acid transporter 1; mTORC1, mechanistic target of rapamycin complex 1; ERβ, estrogen receptor beta; BPA, bisphenol A; BMMFs, bovine meat, and milk factors; DNA, desoxyribonucleic acid; DLBCL, diffuse large B cell lymphoma.
Table 3. Comparison of microRNAs (miRs) implicated in the pathogenesis of DLBCL with bioactive milk-derived microRNAs.
Table 3. Comparison of microRNAs (miRs) implicated in the pathogenesis of DLBCL with bioactive milk-derived microRNAs.
MiRsDLBCL-Related miRsReferencesCow’s Milk-
Derived miRs
References
let-7Over-expression of let-7 suppresses BLIMP1 (PRDM1)[324,325]Highly conserved in milk; component of MDEs[411,427,428,429,430,431,432,433,434]
miR-125bIncreased serum levels of exosomal miR-125b-5p in DLBCL patients associated with shorter progression-free survival time and chemo-therapy resistance;
targets TNFAIP3 (A20), the negative regulator of NF-κB; down-regulates the expression of interferon regulatory factor 4 (IRF4) and BLIMP1; targets TP53; reduces BLIMP1-mediated repression of pri-miR-21
[287,288,326,328,329,330,342]Resistant component of milk EVs and MDEs: Postprandial increase in plasma after consumption of commercial cow’s milk in humans;[392,411,436,446]
miR-21Increased serum levels of patients with DLBCL miR-21; increased in exosome-enriched serum of patients with DLBCL; increased levels in DLBCL tumors; targets MAXD1 (MAX dimerization protein 1) promoting the formation of C-MYC-MAX heterodimers that activate C-MYC expression; targets Von Hippel Lindau mRNA (VHL); inversely correlated with the levels of FOXO1 and PTEN in DLBCL cell lines targets programmed cell death 4 (PDCD4); targets IGFBP3; drives M2 polarization of macrophages[239,286,329,330,331,332,333,334,335,336,337,338,339,343,347,348,349,352,460,462,471,472,473,474]Abundant signature miR of cow’s milk; component of MDEs; targets TNFAIP3, FOXO1, PTEN, BCL11B, and PDCD4; promotes M2 macrophage polarization[238,267,353,354,427,430,446,447,448,449,450,451,452,453,454,455,456,457,473,474,475,476]
miR-29sTransfection of U2932 DLBCL cells twith EBV-encoded latent membrane protein 1 (LMP1) exhibited significant upregulation of miR-29a,b and c; miR-29b enhances RUNX2 expression, which promotes proliferation of B-NHL cells; targets AID (AICDA) enhancing BCL6 expression[496,498,538]Component of MDEs; dose-dependent increase in plasma after intake of commercial cow’s milk; increasing RUNX2 expression in PBMCs[409,411,448]
miR-155Higher levels of miR-155 are present in DLBCL with an ABC phenotype; highly expressed in activated B cells and proliferating lymphoblastoid cell lines; targets the PI3K inhibitor PIK3R1; activates BCR-PI3K-AKT-mTORC1 signaling at various checkpoints via targeting PTEN, DEPTOR, and SHIP1 (INPP5D); EBV upregulates miR-155. Higher miR-155 plasma levels are associated with short survival; higher levels of plasma exosomal miR-155 correlate with reduced R-CHOP response; targets AID (AICDA) enhancing BCL6 expression, the key suppressor of BLIMP1[28,240,241,299,358,359,360,361,362,363,364,365,366,371,372,377,378,535,536,537]Immune-related miR of colostrum and cow’s milk; pretreatment of IEC-6 cells with MDEs increased intracellular levels of miR-155[429,504]
miR-148amiR-148a-3p impairs RAS/ERK signaling in B cells by targeting SOS1/2 thereby reducing ERK-mediated expression of BLIMP1; miR-148a-3p targets DNMT1; enhances the expression of BCL6, the key suppressor of BLIMP1; elevated miR-148a-3p expression impaired B cell tolerance by promoting the survival of immature B cells[28,235,299,380,526,534,539]Signature miR of commercial cow’s milk; component of milk EVs and MDEs; targets DNMT1, TP53, PTEN, SOS1 and SOS2, thereby reduces ERK-mediated activation of BLIMP1; targets MITF, BACH2 and PDL1[384,427,447,511,515,517,526,527,539,551]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Melnik, B.C.; Stadler, R.; Weiskirchen, R.; Leitzmann, C.; Schmitz, G. Potential Pathogenic Impact of Cow’s Milk Consumption and Bovine Milk-Derived Exosomal MicroRNAs in Diffuse Large B-Cell Lymphoma. Int. J. Mol. Sci. 2023, 24, 6102. https://doi.org/10.3390/ijms24076102

AMA Style

Melnik BC, Stadler R, Weiskirchen R, Leitzmann C, Schmitz G. Potential Pathogenic Impact of Cow’s Milk Consumption and Bovine Milk-Derived Exosomal MicroRNAs in Diffuse Large B-Cell Lymphoma. International Journal of Molecular Sciences. 2023; 24(7):6102. https://doi.org/10.3390/ijms24076102

Chicago/Turabian Style

Melnik, Bodo C., Rudolf Stadler, Ralf Weiskirchen, Claus Leitzmann, and Gerd Schmitz. 2023. "Potential Pathogenic Impact of Cow’s Milk Consumption and Bovine Milk-Derived Exosomal MicroRNAs in Diffuse Large B-Cell Lymphoma" International Journal of Molecular Sciences 24, no. 7: 6102. https://doi.org/10.3390/ijms24076102

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop