Next Article in Journal
The VESPA Project: Virtual Reality Interventions for Neurocognitive and Developmental Disorders
Previous Article in Journal
Congenital Epulis: A Two-Case Report
 
 
Journal of Mind and Medical Sciences is published by MDPI from Volume 12 Issue 1 (2025). Previous articles were published by another publisher in Open Access under a CC-BY (or CC-BY-NC-ND) licence, and they are hosted by MDPI on mdpi.com as a courtesy and upon agreement with Valparaiso University (ValpoScholar).
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Involvement of Oxidative Stress in Non-Hodgkin’s Lymphomas; A Review of the Literature

by
Ramona Ingrid Corbeanu
1,2 and
Amelia Maria Găman
2,3,*
1
Doctoral School, University of Medicine and Pharmacy of Craiova, Craiova, Romania
2
Department of Pathophysiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
3
Department of Hematology, Filantropia Hospital of Craiova, Craiova, Romania
*
Author to whom correspondence should be addressed.
J. Mind Med. Sci. 2022, 9(1), 1-15; https://doi.org/10.22543/7674.91.P115
Submission received: 25 September 2021 / Revised: 26 September 2021 / Accepted: 29 November 2021 / Published: 10 April 2022

Abstract

:
Non-Hodgkin’s malignant lymphomas are a heterogeneous group of hematological malignancies, characterized by a variety of clinical, morphological, histopathological, immuno-histochemical, molecular and evolutionary features. They represent a form of cancer that develops from the lymphatic tissue, as a result of the malignant transformation of B (85%) or T (15%) lymphocytes. Lymphomagenesis is described as a multi-stage process involving the mutation and proliferation of cell clones. Oxidative stress is defined as an imbalance of cellular redox status caused by the production of reactive oxygen species (ROS) and/or by decreasing antioxidant systems that allows their accumulation in the cell. Small quantities of ROS are involved in physiological mechanisms such as cell growth and differentiation, cell signaling, antimicrobial defense, phagocytosis. Normally, cells are capable of defending themselves against ROS damage through various scavenger systems. On the other hand, excessive ROS contribute to various diseases such as carcinogenesis, ischemia, atherosclerosis, neurodegenerative diseases. Oxidative stress exerts noxious effects on the cell structures by inducing structural changes in membranes, lipids, proteins or DNA. The present review summarizes the latest findings in understanding the ROS-linked signaling pathways in the initiation of lymphomagenesis, disease progression, metastasis, as well as in the pharmacodynamics of specific treatments for this malignancy.

Introduction

Non-Hodgkin’s lymphoma (NHL) is the most frequent hematological malignancy [1]. The American Cancer Society has estimated 74,200 new cases of NHL for 2019 [2]. The incidence has doubled in the last 50 years, resulting from improved diagnostic techniques and access to medical care. Various classification systems have been developed, but three of them are most commonly used: the National Cancer Institute’s Working Formulation (IWF), the Revised European-American Classification of Lymphoid Neoplasms (REAL) and the World Health Organization (WHO) classification [3,4,5]. The WHO modification of the REAL classification of NHL is based on cell lineage and morphology. In 2016, the World Health Organization’s classification of lymphoid, histiocytic and dendritic neoplasms comprises B-line, T-line or Natural Killer (NK) malignancies, post-transplant lymphoproliferations and histiocytic and dendritic neoplasms, with two sub-divisions: precursor neoplasms and mature differentiated neoplasms [5]. Diagnostic and treatment methods have improved with this revised classification, bringing in the forefront the current cytogenetic and molecular biology data necessary for the most targeted therapeutic management. The natural history of these tumors shows considerable variation. From a clinical-evolutionary point of view, NHL are sorted into three classes: indolent (diffuse lymphocytic B-cell lymphoma, lymphoplasmacytic lymphoma, marginal zone lymphoma, grade I and II follicular lymphoma, Mycosis fungoid), aggressive (B-cell lymphomas: mantle cell lymphoma, grade III follicular lymphoma, large diffuse B-cell lymphoma, primitive mediastinal lymphoma and T-cell lymphomas: angioimmunoblastic, angiocentric, peripheral T-cell, intestinal T-cell, anaplastic with large cells) and very aggressive (lymphoma with B/T precursors, Burkitt lymphoma and other peripheral T-cell lymphomas). In their evolution, indolent lymphomas can progress to aggressive lymphomas. However, there is a paradox, as aggressive lymphomas have a better prognosis than indolent lymphomas [6,7].
Prognosis mainly depends on individual factors (such as age, comorbidities), lymphoma subtype and extent of lymph node involvement. For a better prediction of the outcome and management, the International Prognostic Index (IPI) and its variant were developed [8,9]. In addition to these factors, recent studies emphasize the importance of cytogenetic markers in the prognosis, as well as in the therapeutic strategies of NHL [10].

Discussion

Lymphomagenesis is a complex process resulting from the interactions between genetic and environmental factors. It involves a complex, multi-step process, characterized by a progressive clonal expansion of B-cells or T-cells and/or NK-cells. In the first stage, the proliferation is polyclonal under the action of certain risk factors (Table 1).
Later on, the mutant clone appears as a result of the lesions that affect proto-oncogenes or tumor suppressor genes. The activation of certain oncogenes offers the advantage of autonomous growth and expansion. These oncogenes can be activated by chromosomal translocations or suppressor tumor loci can be inactivated by chromosomal deletions or mutations. Specific chromosomal alterations are associated with significant changes in gene expression. Translocation (14;18)(q32; q21) produces the overexpression of BCL-2 protein which promotes lymphocyte expansion by inhibiting apoptosis. It is associated with follicular lymphoma (FL), but it has also been described in 30-40% cases of diffuse large B-cell lymphoma germinal center B-cell (GCB-DLBCL) [26,27].
The t(8;14)(q24;q32) involving the MYC oncogene which promotes cell proliferation and development is the most common chromosomal abnormality in Burkitt lymphoma (BL) [28]. The t(2;5)(p23;q35) affects the nucleophosmin (NPM) gene and the anaplastic cell lymphoma kinase (ALK) gene which are described in large cell anaplastic lymphomas occurring in 10-20% cases of pediatric lymphomas and in young adults under 30 years of age [29]. The t(11;14)(q13;q32) is present in most cases of mantle cell lymphoma (MCL) with mutant implications in the BCL-1 gene, leading to rapid cell proliferation [30]. Other mechanisms that involve the inactivation of tumor suppressor genes are linked to lymphomagenesis. It is well-known that the tumor protein p53 encoded by the TP53 gene represents a vital tumor suppressor. This activity is fulfilled through DNA repair, apoptosis, senescence, autophagy via transcription-dependent activity (TA) and transcription-independent activity (TIA) in the nucleus and cytoplasm [31]. Normal functions of these pathways are crucial for tumor suppression. Altered lymphocytes undergo p53-dependent apoptosis. Genomic instability caused by the dysfunction of this gene, along with other chromosomal alterations, allows B lymphocytes to escape immune surveillance, having a polyclonal evolution. Therefore, studies have demonstrated that TP53 mutations initiate and maintain the progression of lymphoproliferative disorders [32]. Moreover, TP53 mutations represent an independent prognostic marker of poor survival in DLBCL patients [33].
The purpose of the latest research includes a better understanding of the signaling pathways involved in lymphomagenesis. In the medical practice, most NHLs are of B-line. Proteins that play a key role in these signaling pathways are affected by the chromosomal changes. Recently, two types of aberrant Signal Transducer and Activator of Transcription (STAT) 3 and 5 have been described, as important effectors of cellular transformation and their connection with hematopoietic cancers [34]. Among NHLs, DLBCL is associated with high-level STAT3 expression and activation, especially the activated B-cell like (ABC-DLBCL). Studies have indicated that different subtypes of peripheral T cell lymphomas (PTCL) and NK lymphomas are linked by activating the mutations of STAT 3 and STAT 5B and increased phosphorylated STAT3 and STAT5B proteins which give growth advantage to transduced cell lines or normal NK cells [35]. STAT 5BN642H mutations has been reported in adult T cell-leukemia/lymphoma and γδ-T cell lymphomas, like hepato-splenic TCL, primary cutaneous TCL, monomorphic epitheliotropic intestinal TCL, while STAT 3 domain Y640 F and D661Y/V/H/N mutations have been found in T-large granular lymphocyte leukemia, NK, NK/T and adult T cell leukemia/ lymphoma [36].

What is known about the B cell receptor (BCR) signaling pathways in normal B-lymphocytes?

BCR is a signaling complex expressed by the most normal and malignant B lymphocytes. It is a transmembrane signaling complex involved in the proliferation, differentiation, adhesion or apoptosis of these cells [37,38]. Initially, it was considered that an inducible loss of murine BCR causes the death of peripheral B-cells. Subsequently, it was concluded that both ligand-independent activation in the absence of the receptor and the activation sustained by its presence are important in normal B-cell survival. It is thought that the chronic activation of the BCR pathways is responsible for various B-cell malignancies. Studies on B-line lymphomas, such as DLBCL, FL, MCL, and BL have shown major importance in the survival and proliferation of lymphocytes through BCR [39]. Receptor activation is dependent on nuclear factor Kappa-B (NF-kB) and PI3K mediated signaling pathways. Normally, NF-kB is involved in several stages of growth and differentiation of B and T lymphocytes, having a protective role on lymphocyte precursors, ensuring an anti-apoptotic role against tumor necrotic factors (TNF-α) [40]. Constitutional mutations of BCR at immunoglobulins α and β levels have been identified in primary DLBCL, especially in ABC-DLBCL expressing high levels of NF-kB activity and associated with a poor outcome [41]. These mutations are not the only ones which increase the cellular response to BCR activation. Studies demonstrate the existence of additional mutations, such as CARD11 mutation which ensures the activation and potentiates NF-kB activity [42,43].
Many studies have evaluated the role of Wnt signaling pathways in carcinogenesis [44,45]. Normally, this signaling pathway is involved in cell proliferation, differentiation, survival and apoptosis, as well as, angiogenesis. The Wnt signaling pathway is characterized as canonical and non-canonical, B-catenin-dependent and B-catenin-independent. In the absence of the ligand, B-catenin is destroyed by a complex. It binds to receptor-transmembrane frizzled proteins (Fz proteins) and low-density lipoprotein receptor-related protein 5/6 (LRP 5/6) and reaches the nucleus. Through T-cell factor and lymphoid-enhancing factor (Tcf/Lef), transcription factors produce cellular changes. Several studies have documented its involvement in carcinogenesis, leukemogenesis, as well as in lymphomagenesis (DLBCL, BL, and MCL) [46,47,48].
For a better understanding of the molecular mechanisms underlying the pathogenesis of lymphomas, the researchers studied how signaling pathways potentiate one other. MiR-101 is associated with proliferation, migration, invasion and cell apoptosis [49,50]. Its role in carcinogenesis was demonstrated by various studies [51,52]. Recently, Huang et al. developed a theory according to which miR-101 expression is down-regulated in DLBCL [53]. Therefore, low levels of miR-101 in patients with DLBCL are correlated with tumor progression by targeting KDM1a pathway.

The involvement of oxidative stress in NHL

Involvement of oxidative stress in lymphomagenesis has been the main aim of several studies [54,55,56,57]. Oxygen participates in energy production, a process that takes place in the mitochondria. Following this process, intermediate forms of reactive oxygen (ROS) and nitrogen species (RNS) appear into the body within the basal cell metabolism, but also as a result of the exposure to exogenous factors such as pollution, cigarette smoke or hyperoxia [58]. Numerous enzymes participate in the process of producing free radicals (FR), including nicotinamide-adenine-dinucleotide phosphate (NADPH), succinate dehydrogenase (SDH), cytochrome c reductase, cytochrome b5, monoamine oxidases (MAO-A and MAO-B), pyruvate dehydrogenase complex (PGDH). A variable number of endogenous antioxidant defense systems have the ability of neutralizing ROS. These antioxidant systems include enzymes and non-enzymatic systems [59,60,61]. Oxidative stress is a biochemical imbalance of cellular redox status caused by the production of reactive oxygen species and/or by decreasing the antioxidant systems that allow their accumulation in the cell [62]. Small quantities of ROS intervene in the physiological mechanisms, such as cell growth and differentiation, cell signaling, antimicrobial defense, phagocytosis. In exchange, large quantities produce pathological processes such as inflammation, carcinogenesis, ischemia, atherosclerosis, neurodegenerative diseases and allergies [63,64,65,66].
It is considered that inflammation is a protective mechanism of the body against cell destruction, but there is a correlation between NHL, inflammation and oxidative stress. Sustained antigenic stimulation promotes the clonal proliferation of lymphocytes by inhibiting apoptosis and the activation of NF-kB by pro-inflammatory cytokines and growth factors [67]. Clonal proliferation is also supported by elevated levels of B lymphocyte growth factor (BAFF/BlyS) as shown in some autoimmune disorders, but also in FL [68]. Oxidative stress can activate a variety of transcription factors that lead to the expression of the genes involved in chronic inflammation, increase the level of pro-inflammatory cytokines (mainly interleukin-1), stimulate and activate B lymphocytes to produce antibodies and alter cellular DNA [69]. Recent studies have shown that genetic variations of pro-inflammatory cytokines (TNF-α, interleukin-10) double the risk of DLBCL, a subtype particularly associated with autoimmune diseases, as demonstrated by other studies [70]. Genomic instability in B lymphocytes, as well as the activation of a transcription factor, NF-kB, give an advantage of autonomous growth and expansion to mutant lymphocytes [71]. Therefore, chronic infection disrupts cell growth and survival [72].
Genes controlling the redox homeostasis are important ‘actors’ in lymphomagenesis, therefore in a multi-center study, performed on 1,172 cases of NHL, ten oxidative stress genes (AKR1A1, AKR1C1, GPX, MPO, NOS2A, NOS3, OGG1, PPARG, SOD2, CYBA) were analyzed and it was established that the genetic variations of these genes lead to a high status of ROS, thus increasing the risk of NHL, especially DLBCL, one of the most common and aggressive subtypes of NHL [73]. Based on this idea, subsequent studies conducted on the Korean population group highlighted the link between the gene polymorphism involved in the DNA repair and the risk of NHL, as follows: four gene genotypes (XRCC1 399 GA, OGG1 326 GG, BRCA1 871 TT and WRN 787 TT) have a low risk of NHL, while MGMT 115 CT genotype is associated with an increased risk. Regarding the MDR1 gene (multidrug resistance 1), genotype 1236 CC is associated with a low risk, while genotypes 3435 CT and TT have an increased risk of NHL [74].
Oxidative stress defined as an imbalance between oxidants and antioxidants, in favor of the oxidants, destroys nuclear and mitochondrial DNA [75] and favors the appearance of specific markers such as 8-hydroxy-2-deoxyguanosine (8-OHdG) [76], protein carbonyl groups as a marker of protein oxidation [77], malondialdehyde (MDA) and F2-isoprostanes as markers of lipid peroxidation [78,79]. Mezayen et al. evaluated the activity of oxidant/antioxidant status in patients with NHL, before and one month after the specific cytotoxic regimen and concluded that there was an increase in MDA and a reduction in SOD levels, thus suggesting that chemotherapy destroys the balance [80]. High-levels of 8-OHdG in tumors, blood samples and urine represent a promising marker for predicting the prognosis of cancers [81,82,83,84]. Similarly, other scientists have shown a correlation between elevated 8-OHdG levels and an increased risk of developing malignant hematological diseases [85] and highlighted that elevated urinary levels of 8-OHdG are associated with a poor prognosis in patients with FL [86]. Guanosine hydroxylation is the result of normal metabolism processes and environmental factors, such as exposure to cigarette smoke, asbestos, heavy metals or polycyclic aromatic hydrocarbons. For this purpose, a study conducted by Fenga et al. in 2017 demonstrated that 8-OHdG can be used as a non-invasive marker of early genotoxic DNA damage following the exposure to low doses of benzene [87]. The conclusion was that benzene metabolism (through cytochrome P450) induces FR that affect the oxidant/antioxidant balance, increases the level of ROS and produces increased toxicity, affecting cell proliferation, differentiation and apoptosis (through p38-MAPK signaling pathways, SAPK/JNK, STAT3).
Oxidative stress is involved in various stages of the cell cycle, by activating cell signaling pathways, including tumor cell proliferation, migration and increased tumor cell pro-angiogenic factors. DNA-oxidative damage via moderate levels of ROS lead to several repercussions: it causes genomic instability, favors BCR and oncogenic kinase signaling, promotes B-cell survival and facilitates tumor progression [88]. On the contrary, high levels of ROS produce cell death. Eliminating cellular mechanisms that maintain efficient ROS levels might turn the balance of protumorigenic ROS activity toward cancer cell death. The role of antioxidant systems in carcinogenesis has been debated over the years. However, it has now been established that exaggerated antioxidant responses, regulated by specific signaling pathways, have an important impact in lymphomas. For example, various studies demonstrated that there is a correlation between rs1001179 polymorphisms and lower catalase activity which interferes with the response to oxidative stress and enhances tumorigenesis [89,90]. Catalase is an important endogenous antioxidant system that decomposes H₂O₂ into oxygen and water. In 2017, Wang et al. conducted a meta-analysis which studied the correlation between GPX-1 and cancer risk [91]. Their conclusion was that there are specific polymorphisms of this enzyme family, especially for patients with TT/CT genotype who have an increased risk of bladder cancer and brain tumors, but no evidence was found between these polymorphisms and NHL.
It is known that uric acid is the final product of purine metabolism, important components of nucleic acids and coenzymes that can be synthesized in the body or can be obtained by eating certain foods. Over the years, arguments have been brought to support the role of uric acid as an antioxidant [92,93]. This fundamental idea is supported by another study which has suggested that uric acid improves indomethacin-induced enteropathy in mice through its antioxidant effect [94]. A study conducted in 2015 assigned the link between p53 and a uric acid transporter – SLC2A9 or GLUT9 – which functions as an antioxidant, capable of protecting cells against ROS [95].
Increased ROS levels in certain tumor cells due to metabolic changes or classical chemotherapeutic use, rely on targeting specific antioxidant pathways. Thioredoxin (Trx) and GSH represent two antioxidant systems with an important role in the regulation of cellular redox homeostasis. The researchers have tried to utilize the pharmacological inhibition of antioxidant enzymes or other ROS-inducing molecules to describe the specific antioxidant defense, but with limited efficacy in monotherapy regimens. Trx family members represent a major antioxidant system, containing Trx 1 protein, thioredoxin reductase (TrxR) and NADPH. For instance, in BL cell line models two specific compounds were utilized, i.e. SK053 and adenanthin, with specific targets on Trx, TrxR and periredoxins (Prx) 1 and 2, two H₂O₂-scavenging enzymes [96,97]. These molecules trigger ROS-induced extracellular signal-regulated kinase ½ (ERK1/2) activation and induce apoptosis. Auranofin, a gold complex, is the inhibitor of TrxR with antitumor activity in the preclinical models of chronic lymphocytic leukemia [98] and classical Hodgkin lymphoma [99]. Regarding, G.S.H., elevated levels are associated with different types of cancer and chemoresistance [100]. Therefore, pharmacologists paid attention to a GSH-depleting agent, buthionine sulfoximine against cancer progression and chemoresistance, which is more effective in combinations with other therapeutic drugs [101].

The main subtypes and their specific features related to oxidative stress

Diffuse large B-cell lymphoma is the most frequent aggressive B-cell NHL [102]. Flow-cytometry identifies typically B-cell antigens like CD19, CD20, CD22, CD79a and CD45. Molecular features have a prognostic impact. C-MYC is a proto-oncogene on the chromosome 8q24, which confers the advantage of proliferation and growth of malignant cells when deregulated. Translocation (14;18) is associated with the overexpression of BCL-2, an antiapoptotic factor, which along with c-MYC, characterizes double-expresser lymphoma (DEL), with intermediate prognosis. When the deregulation of BCL-6 oncogene is added, the DLBCL turns to aggressive triple-hit lymphoma [103]. Four distinct genetic subtypes of the disease with recurring mutation have been recently identified by Schmitz and colleagues: MCD, BN2, N1 and EZB [104]. The MCD subtype is characterized by the presence of MYD88 and CD79 mutations, the N1 has NOTCH1 mutations, the BN2 by BCL-6 and NOTCH2 mutations and the EZB subtype has EZH2 and BCL2 translocations. These facts have an important clinical outcome, the BN2 and EZB subtype have good prognoses. Genetic mutations are not the only ones responsible for malignant evolution. As demonstrated, oxidative stress can initiate lymphomagenesis, but also the progression of the disease. A study that evaluated oxidative stress in DLBCL patients via the Free Oxygen Radical Testing (FORT) for FR and via the Free Oxygen Radical Defense (FORD) for the antioxidant status has concluded that patients with advanced stage DLBCL have a higher level of FORT and a decreased level of FORD which demonstrates that ROS are involved in NHL pathogenesis [72]. Tumors in patients with advanced disease and poor prognosis have deregulated the expression of certain ROS-metabolizing enzymes and extended pro-oxidant phenotype. According with these findings, Kinowaki et al. demonstrated the link between the overexpression of GPX4 in DLBCL and worse prognosis [105]. GPX4 is an intracellular antioxidant enzyme which prevents ROS-induced cell death. Therefore, an antioxidant treatment of advanced tumors may help patients manage oxidative stress conditions. The Trx system defends both normal and malignant cells against oxidative stress [106]. However, the overexpression of Trx-1 gives cancer cells the advantage of growth, proliferation, survival and it also correlates with drug resistance, as reported in DLBCL-derived cell lines when compared to normal B cells, using Western blotting and real-time PCR [107]. But, the latest studies have shown a new possible hope by using pro-oxidant therapies to urge cancer cells toward death with promising aspects in B-cell malignancies. Therefore, Wang and colleagues have established that the down-regulation of Trx-1 sensitized lymphoma cells to chemotherapy regimens [108]. According to this idea, cells lacking Trx are more sensitive to ROS due to reduced proapoptotic activity of Forkhead box protein O1 (FOXO1) [109]. Trx reduces the bonds between FOXO1 and p300, formed as a response to oxidative stress. The depletion of Trx facilitated p300-mediated acetylation of FOXO1 and mediated cell death. These findings underline the role of Trx in the pathogenesis of DLBCL and are a solid argument for therapeutic exploitation. Recent studies have evaluated the involvement of the second group members thioredoxin-domain-containing (TXNDC) proteins 2,3 and 6 in DLBCL and demonstrate that these proteins are all expressed in testicular and systemic lymphomas with clinical importance [110]. This study included a limited number of cases (28 de novo confirmed systemic DLBCL and 21 testicular DLBCL) so, further investigations are needed. An interesting drug-resistance mechanism involving antioxidant enzymes was reported in ABC-DLBCL, with chronic activation BCR signaling. In these DLBCLs, the activity of STAT3 led to the upregulation of antioxidant enzyme SOD2 mitochondrial and mediated doxorubicin resistance. The use of STAT3 inhibitor oppressed SOD2-dependent resistance mechanisms and restored doxorubicin sensitivity in preclinical models [111].
Grade I and II follicular lymphoma is an indolent B cell lymphoproliferative disorder of transformed follicular center B cells, while grade III is an aggressive NHL. In, F.L., 85% of the cases have t(14;18) which results in the overexpression of the BCL-2 protein. Recent studies have developed a theory according to which this translocation can also be identified in normal cells, so it is considered that additional mutations are needed to produce neoplasia, such as the methyl transferase H3K27 EZH2 mutation, recognized in 27% of FL [112]. There is a particular subtype of FL, the pediatric form, which does not describe the BCL-2 rearrangement and which, according to the 2016 WHO classification, has a high possibility of curability [113]. The involvement of oxidative stress in this subtype has recently been demonstrated [84]. Peroja and colleagues evaluated redox-state regulating enzymes Prx I-VI and Trx in untreated FL [114]. In a cohort of 76 histologically confirmed FLs, the expression of Prx I-VI, Trx and the oxidative stress marker, nitrotyrosine, were assessed. The immuno-histochemical results were correlated with clinical prognostic factors such as disease-specific survival, progression-free survival and overall survival. Their findings suggested that high Prx levels have a good disease-specific survival and overall survival which exhibit a protective role in FL patients.
Translocation (11;14) is present in most cases of mantle cell lymphoma with mutant implications in the BCL-1 gene. There are also forms in the absence of cyclin D1, but the overexpression of cyclin D2, with a better prognosis, is independent of the risk factors [115]. Zhang and colleagues correlated B-cell-specific transcription factor (BACH2), a tumor suppressor factor with hypoxic environment in MCL [116]. Their explanation lies in the fact that oxygen depletion produces an increased level of ROS and excessive heme, which are harmful to the body. Excess heme causes BACH2 degradation in B lymphocytes. Under hypoxic conditions, HIF-1α independently produces the downregulation of BACH2. Under these aspects, the survival of cancer cells in MCL is ensured by the downregulation of BACH [117].
Mucosa-associated lymphoid tissue lymphoma is an indolent, multifocal lymphoma. Between 4-20% are gastric MALT lymphomas, strongly associated with the HP infection. There is a loop between HP infection-chronic inflammation-ROS release and lymphomagenesis initiation [118]. The malignant transformation of B lymphocytes occurs as a result of t(11;18), (1;14), (14;18) during the HP infection, via the activation of NF-kB [119]. If the tumor’s proliferation is limited to the mucosa and it is closely related to the infectious agent, sometimes it is sufficient to eradicate the infection by means of antibiotic therapy in order to stop lymphomatous proliferation. Studies have shown that t(11;18) is associated with antibiotic resistance and a worse prognosis [120].
Small lymphocytic lymphoma is a neoplasm of mature clonal B lymphocytes with a typical immunophenotype expression of CD5 and CD23 surface antigen and mutated variable regions of the immunoglobulin heavy-chain (IGHV) genes in 50-70% of the cases [121]. The genomic background provides specific chromosomal abnormalities with prognostic importance, such as: 17p or 11q deletion indicate poor prognosis, whereas 13q deletion represents a better prognosis [122]. The role of oxidative stress in the pathogenesis of this malignancy has been discussed in several studies that have shown correlations between the level of oxidative stress markers and cytogenetic changes [123,124]. Supporting this idea, some researchers discovered that high levels of MDA, protein carbonyl and decreased levels of antioxidants (SOD and catalase) were associated with 17p deletion and worse clinical presentation [125].

Oxidative stress and NHL management

The most important findings were pointed out on the involvement of oxidative stress in the initiation and progression of NHL, but it seems that it also intervenes in the mechanisms of action of some cytostatic agents. Chemotherapeutic regimens included drugs that destroy cancer cells through different mechanisms. One possible mechanism is related to the redox cellular status. The adjustment of oxidative stress is a relevant factor in both tumor development and the responses to anticancer therapies. As seen before, ROS can stimulate tumor formation by inducing DNA mutations and pro-oncogenic signaling pathways. On the other hand, high ROS levels are usually harmful to cells and may represent a barrier for tumorigenesis. A large number of chemotherapeutic drugs exert their activity through high levels of ROS by interfering with antioxidant systems or ROS inducing pathways [126]. Cyclophosphamide is an alkylating agent used in the treatment of various cancers [127] which generates cell death through its 2 metabolites, acrolein and phosporamide [128]. Acrolein is responsible for the decreased overall antioxidant capacity and the activation of the stress-signaling MAP-kinases JNK, p42/44 and p38 pathways with an increased production of ROS and subsequently oxidative stress causing DNA damage, lipid peroxidation, increased permeability of the mitochondrial membrane, release of cytochrome C, activation of caspase-3 and apoptotic cell death [129]. Studies have shown that doxorubicin causes cell death by increasing ROS levels via p53-dependent manner, but it also promotes the accumulation of a transcription factor, FOXO3, which activates two apoptotic genes (Noxa and BIM) with subsequent release of cytochrome C and ROS [130]. The anti-CD20 antibody, rituximab, has a proapoptotic effect by downregulating BCL-2 (an antiapoptotic factor) and inhibiting survival p38 MAPK signal pathways. Its cytotoxic effect is also due to a complement-dependent process that generates increased amounts of ROS [131]. Glucocorticoids have a considerable effect on oxidative stress which depends on the duration of the treatment. These hormones increase the metabolic rate, mitochondrial membrane potential and mitochondrial oxidation, which in turn increases the production of superoxide, H₂O₂ and hydroxyl radicals leading to a state of cellular oxidative stress which causes oxidative damage to the DNA, protein carbonyl formation and membrane lipid peroxidation [132]. The benefits of radiotherapy (RT) for lymphomas have been well documented for many years. The RT concept is based on the alteration of DNA cancer cells, which can be done directly and indirectly [133]. The appearance of ROS is incriminated for indirect DNA damage, both in cancer cells exposed to the area of irradiation and in healthy cells at a distance from it. The penetration of heavy particles of ionizing radiation in the body produces the breakdown of chemical bonds and, on the other hand, the water which represents half of the body weight, is split by ionizing radiation, these mechanisms favoring the assimilation of hydroxyl ROS. However, there is a thin line between the useful level and the harmful level of oxidative stress in cancer treatments. When pro-oxidant treatment is used, malignant cells generate DNA oxidation, proteins oxidation and lipid peroxidation being the cause of the increased tumor burden. Some of the antioxidants, such as vitamin C, can act both as antioxidants and pro-oxidants [134]. The pro-oxidant effect of vitamin C was observed at high doses, having an anticancer role. Ascorbate produces large amounts of H₂O₂ which activates proapoptotic signaling pathways, causes DNA damage and ultimately, cell death. Further evidence has been provided that targeting the antioxidant capacity of tumor cells can have a reliable therapeutic impact [135,136].
Nowadays, antioxidants are in the spotlight as an emerging solution to various disorders, such as cardio-metabolic disorders, solid and blood cancers via reducing the oxidative stress and the inflammation levels, e.g., increasing TAC and SOD and decreasing MDA, TNF-α and C-reactive protein concentrations, and pathogenic links between the antioxidant levels in the serum and the development of malignancies has been demonstrated [137,138,139,140,141]. To this regard, recent studies have focused on the effect of melatonin (MLT) in different diseases [142]. MLT is a natural hormone of the pineal gland. MLT activates MT1 and MT2 receptors and takes part in learning, memory and neuroprotection [143]. Besides its role in maintaining our wake-sleep cycle, melatonin may have anticancer activity through antiproliferative, antioxidant and immunostimulant effects via different pathways [144,145].
Yan et al. demonstrated anti-tumor activity in Hodgkin’s lymphoma by inhibiting cell proliferation and by promoting apoptosis via the increased expression of LC3-II and the decreased p62 proteins [146]. Knowing its anti-inflammatory and anti-oxidant effects, it was concluded that this molecule may represent an adjuvant treatment in several types of cancer. The beneficial effect has been reflected both in the quality of life and control over the disease (growth, size, effectiveness of chemotherapies) [147,148]. The data coming from our research group which evaluated the involvement of oxidative stress in lymphoproliferative, e.g., chronic lymphocytic leukemia and DLBCL, and myeloproliferative neoplasms, e.g., essential thrombocythemia, pointed out that the aforementioned malignancies are associated with increased reactive oxygen species levels and reduced antioxidant capacity [149,150,151,152].

Highlights

Lymphomagenesis is associated with increased oxidative stress levels.
Oxidative stress can also be involved in the action of several chemotherapeutic drugs.
The involvement of oxidative stress has been studied in DLBCL, FL, MCL, MALT and small lymphocytic lymphomas.

Conclusions

The current evidence suggests that oxidative stress is involved in lymphomagenesis, genetic instability, disease progression and NHL management. Further studies are therefore necessary to evaluate if and how oxidative stress-modulating therapies can influence these processes.

Conflicts of Interest disclosure

There are no known conflicts of interest in the publication of this article. The manuscript was read and approved by all authors.

Compliance with ethical standards

Any aspect of the work covered in this manuscript has been conducted with the ethical approval of all relevant bodies and that such approvals are acknowledged within the manuscript.

References

  1. Chihara, D.R.; Nastoupil, L.J.; Williams, J.N.; Lee, P.; Koff, J.L.; Flowers, C.R. New insights into the epidemiology of non-Hodgkin lymphoma and implicatios for therapy. Expert Rev Anticancer Ther. 2015, 15, 531–544. [Google Scholar] [CrossRef] [PubMed]
  2. American Cancer Society. Cancer facts and figures [online]. Available online: https://www.cancer.org/content/dam/cancer-org/research/cancer-facts-and-statistics/annual-cancer-facts-and-figures/2019/cancer-facts-and-figures-2019.pdf (accessed on 29 May 2019).
  3. National Cancer Institute sponsored study of classifications of non-Hodgkin’s lymphomas: summary and description of a working formulation for clinical usage. The Non-Hodgkin’s Lymphoma Pathologic Classification Project. Cancer. 1982, 49, 2112–2135. [Google Scholar] [CrossRef]
  4. Harris, N.L.; Jaffe, E.S.; Stein, H.; Banks, P.M.; Chan, J.K.; Cleary, M.L.; Delsol, G.; De Wolf-Peeters, C.; Falini, B.; Gatter, K.C.; et al. A revised European-American classification of lymphoid neoplasms: a proposal from the International Lymphoma Study Group. Blood. 1994, 84, 1361–1392. [Google Scholar]
  5. 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.; Jaffe, E.S. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood. 2016, 127, 2375–2390. [Google Scholar] [CrossRef]
  6. Ansell, S.M. Non-Hodgkin lymphoma: diagnosis and treatment. Mayo Clin Proc. 2015, 90, 1152–1163. [Google Scholar] [CrossRef]
  7. Connors, J.M. Non-Hodgkin lymphoma: the clinician’s perspective--a view from the receiving end. Mod Pathol. 2013, 26 (Suppl 1), S111–S118. [Google Scholar] [CrossRef]
  8. Schmitz, N.; Zeynalova, S.; Nickelsen, M.; Kansara, R.; Villa, D.; Sehn, L.H.; Glass, B.; Scott, D.W.; Gascoyne, R.D.; Connors, J.M.; Ziepert, M.; Pfreundschuh, M.; Loeffler, M.; Savage, K.J. CNS International Prognostic Index: A Risk Model for CNS Relapse in Patients With Diffuse Large B-Cell Lymphoma Treated With R-CHOP. J Clin Oncol. 2016, 34, 3150–3156. [Google Scholar] [CrossRef]
  9. Freedman, A.; Jacobsen, E. Follicular lymphoma: 2020 update on diagnosis and management. Am J Hematol. 2020, 95, 316–327. [Google Scholar] [CrossRef]
  10. Antoine-Poirel, H.; Heimann, P. Cytogenetic and molecular testing in lymphoma patients. Belg J Hematol. 2018, 9, 266–278. [Google Scholar]
  11. Velavan, T.P. Epstein-Barr virus, malaria and endemic Burkitt lymphoma. EBioMedicine. 2019, 39, 13–14. [Google Scholar] [CrossRef]
  12. Yamagishi, M.; Fujikawa, D.; Watanabe, T.; Uchimaru, K. HTLV-1-Mediated Epigenetic Pathway to Adult T-Cell Leukemia-Lymphoma. Front Microbiol. 2018, 9, 1686. [Google Scholar] [CrossRef] [PubMed]
  13. Cesarman, E.; Damania, B.; Krown, S.E.; Martin, J.; Bower, M.; Whitby, D. Kaposi sarcoma. Nat Rev Dis Primers. 2019, 5, 9. [Google Scholar] [CrossRef] [PubMed]
  14. Taborelli, M.; Polesel, J.; Montella, M.; Libra, M.; Tedeschi, R.; Battiston, M.; Spina, M.; Di Raimondo, F.; Pinto, A.; Crispo, A.; Grimaldi, M.; Franceschi, S.; Dal Maso, L.; Serraino, D. Hepatitis B and C viruses and risk of non-Hodgkin lymphoma: a case-control study in Italy. Infect Agent Cancer 2016, 11, 27. [Google Scholar] [CrossRef]
  15. Salar, A. Gastric MALT lymphoma and Helicobacter Pylori. Medicina Clinica (Barcelona). 2019, 152, 65–71. [Google Scholar] [CrossRef]
  16. Koller, M.C.; Aigelsreiter, A. Chlamydia psittaci in ocular adnexal MALT lymphoma: a possible causative agent in the pathogenesis of this disease. Current Clinical Microbiology Reports. 2018, 5, 261–267. [Google Scholar] [CrossRef]
  17. Remy, R.; Sylvain, C.; Bachy, E.; Bardel, E. Chronic Borrelia Burgdorferi infection triggers NKT lymphomagenenis. Blood. 2018, 132, 2691–2695. [Google Scholar] [CrossRef]
  18. Melenotte, C.; Mezouar, S.; Mege, J.L.; Gorvel, J.P.; Kroemer, G.; Raoult, D. Bacterial infection and non-Hodgkin’s lymphoma. Crit Rev Microbiol. 2020, 46, 270–287. [Google Scholar] [CrossRef]
  19. Re, A.; Cattaneo, C.; Rossi, G. Hiv and Lymphoma: from Epidemiology to Clinical Management. Mediterr J Hematol Infect Dis. 2019, 11, e2019004. [Google Scholar] [CrossRef]
  20. Travaglino, A.; Pace, M.; Varricchio, S.; Insabato, L.; Giordano, C.; Picardi, M.; Pane, F.; Staibano, S.; Mascolo, M. Hashimoto Thyroiditis in Primary Thyroid Non-Hodgkin Lymphoma. Am J Clin Pathol. 2020, 153, 156–164. [Google Scholar] [CrossRef]
  21. Dopart, P.J.; Locke, S.J.; Cocco, P.; Bassig, B.A.; Josse, P.R.; Stewart, P.A.; Purdue, M.P.; Lan, Q.; Rothman, N.; Friesen, M.C. Estimation of Source-Specific Occupational Benzene Exposure in a Population-Based Case-Control Study of Non-Hodgkin Lymphoma. Ann Work Expo Health. 2019, 63, 842–855. [Google Scholar] [CrossRef]
  22. Qin, L.; Deng, H.Y.; Chen, S.J.; Wei, W. A Meta-Analysis on the Relationship Between Hair Dye and the Incidence of Non-Hodgkin’s Lymphoma. Med Princ Pract. 2019, 28, 222–230. [Google Scholar] [CrossRef] [PubMed]
  23. Zhang, L.; Rana, I.; Shaffer, R.M.; Taioli, E.; Sheppard, L. Exposure to glyphosate-based herbicides and risk for non-Hodgkin lymphoma: A meta-analysis and supporting evidence. Mutat Res Rev Mutat Res. 2019, 781, 186–206. [Google Scholar] [CrossRef] [PubMed]
  24. Hu, L.; Luo, D.; Zhou, T.; Tao, Y.; Feng, J.; Mei, S. The association between non-Hodgkin lymphoma and organophosphate pesticides exposure: A meta-analysis. Environ Pollut. 2017, 231 Pt 1, 319–328. [Google Scholar] [CrossRef]
  25. Rhoades, M.G.; Meza, J.L.; Beseler, C.L.; Shea, P.J.; Kahle, A.; Vose, J.M.; Eskridge, K.M.; Spalding, R.F. Atrazine and nitrate in public drinking water supplies and non-hodgkin lymphoma in nebraska, USA. Environ Health Insights. 2013, 7, 15–27. [Google Scholar] [CrossRef]
  26. Ma, E.S. Recurrent Cytogenetic Abnormalities in Non-Hodgkin’s Lymphoma and Chronic Lymphocytic Leukemia. Methods Mol Biol. 2017, 1541, 279–293. [Google Scholar] [CrossRef]
  27. Pasqualucci, L.; Dalla-Favera, R. Genetics of diffuse large B-cell lymphoma. Blood. 2018, 131, 2307–2319. [Google Scholar] [CrossRef]
  28. López, C.; Kleinheinz, K.; Aukema, S.M.; et al. Genomic and transcriptomic changes complement each other in the pathogenesis of sporadic Burkitt lymphoma. Nat Commun. 2019, 10, 1459. [Google Scholar] [CrossRef]
  29. Kourentzi, K.; Crum, M.; Patil, U.; Prebisch, A.; Chavan, D.; Vu, B.; Zeng, Z.; Litvinov, D.; Zu, Y.; Willson, R.C. Recombinant expression, characterization, and quantification in human cancer cell lines of the Anaplastic Large-Cell Lymphoma-characteristic NPM-ALK fusion protein. Sci Rep. 2020, 10, 5078. [Google Scholar] [CrossRef]
  30. Jain, A.G.; Chang, C.C.; Ahmad, S.; Mori, S. Leukemic Non-nodal Mantle Cell Lymphoma: Diagnosis and Treatment. Curr Treat Options Oncol. 2019, 20, 85. [Google Scholar] [CrossRef]
  31. Candi, E.; Agostini, M.; Melino, G.; Bernassola, F. How the TP53 family proteins TP63 and TP73 contribute to tumorigenesis: regulators and effectors. Hum Mutat. 2014, 35, 702–714. [Google Scholar] [CrossRef]
  32. Voropaeva, E.N.; Pospelova, T.I.; Voevoda, M.I.; Maksimov, V.N.; Orlov, Y.L.; Seregina, O.B. Clinical aspects of TP53 gene inactivation in diffuse large B-cell lymphoma. BMC Med Genomics. 2019, 12 (Suppl 2). [Google Scholar] [CrossRef]
  33. Chen, P.T.; Jorsan, K.; Avezbakiyev, B.; Akhtar, C.; Wang, J.C. Aggressive Diffuse Intermediate Size B-Cell Lymphoma With P53 Mutation Presented as Primary Bone Marrow Lymphoma. J Investig Med High Impact Case Rep. 2020, 8, 2324709620982765. [Google Scholar] [CrossRef] [PubMed]
  34. Brachet-Botineau, M.; Polomski, M.; Heidi, A.N.; Juen, L.; Hédou, D.; Viaud-Massuard, M.C.; Prié, G.; Gouilleux, F. Pharmacological Inhibition of Oncogenic STAT3 and STAT5 Signaling in Hematopoietic Cancers. Cancer. 2020, 12, 240. [Google Scholar] [CrossRef] [PubMed]
  35. Küçük, C.; Jiang, B.; Hu, X.; et al. Activating mutations of STAT5B and STAT3 in lymphomas derived from γδ-T or NK cells. Nat Commun. 2015, 6, 6025. [Google Scholar] [CrossRef]
  36. Waldmann, T.A.; Chen, J. Disorders of the JAK/STAT Pathway in T Cell Lymphoma Pathogenesis: Implications for Immunotherapy. Annu Rev Immunol. 2017, 35, 533–550. [Google Scholar] [CrossRef]
  37. Gomes de Castro, M.A.; Wildhagen, H.; Sograte-Idrissi, S.; Hitzing, C.; Binder, M.; Trepel, M.; Engels, N.; Opazo, F. Differential organization of tonic and chronic B cell antigen receptors in the plasma membrane. Nat Commun. 2019, 10, 820. [Google Scholar] [CrossRef]
  38. Lee, J.; Sengupta, P.; Brzostowski, J.; Lippincott-Schwartz, J.; Pierce, S.K. The nanoscale spatial organization of B-cell receptors on immunoglobulin M-and G-expressing human B-cells. Mol Biol Cell. 2017, 28, 511–523. [Google Scholar] [CrossRef]
  39. Efremov, D.G.; Turkalj, S.; Laurenti, L. Mechanisms of B cell receptor activation and responses to B cell receptor inhibitors in B cell malignancies. Cancers. 2020, 12, 1396. [Google Scholar] [CrossRef]
  40. Derudder, E.; Herzog, S.; Labi, V.; Yasuda, T.; Köchert, K.; Janz, M.; Villunger, A.; Schmidt-Supprian, M.; Rajewsky, K. Canonical NF-κB signaling is uniquely required for the long-term persistence of functional mature B cells. Proc Natl Acad Sci U S A. 2016, 113, 5065–5070. [Google Scholar] [CrossRef]
  41. Lenz, G. Novel NF-κB regulator in ABC DLBCL. Blood. 2016, 127, 2785–2786. [Google Scholar] [CrossRef]
  42. Jattani, R.P.; Tritapoe, J.M.; Pomerantz, J.L. Intramolecular Interactions and Regulation of Cofactor Binding by the Four Repressive Elements in the Caspase Recruitment Domain-containing Protein 11 (CARD11) Inhibitory Domain. J Biol Chem. 2016, 291, 8338–8348. [Google Scholar] [CrossRef] [PubMed]
  43. Wang, Z.; Hutcherson, S.M.; Yang, C.; Jattani, R.P.; Tritapoe, J.M.; Yang, Y.K.; Pomerantz, J.L. Coordinated regulation of scaffold opening and enzymatic activity during CARD11 signaling. J Biol Chem. 2019, 294, 14648–14660. [Google Scholar] [CrossRef] [PubMed]
  44. Janovská, P.; Bryja, V. Wnt signalling pathways in chronic lymphocytic leukaemia and B-cell lymphomas. Br J Pharmacol. 2017, 174, 4701–4715. [Google Scholar] [CrossRef] [PubMed]
  45. Frenquelli, M.; Tonon, G. WNT Signaling in Hematological Malignancies. Front Oncol. 2020, 10, 615190. [Google Scholar] [CrossRef]
  46. Mathur, R.; Sehgal, L.; Braun, F.K.; Berkova, Z.; Romaguerra, J.; Wang, M.; Rodriguez, M.A.; Fayad, L.; Neelapu, S.S.; Samaniego, F. Targeting Wnt pathway in mantle cell lymphoma-initiating cells. J Hematol Oncol. 2015, 8, 63. [Google Scholar] [CrossRef]
  47. Undi, R.B.; Gutti, U.; Sahu, I.; Sarvothaman, S.; Pasupuleti, S.R.; Kandi, R.; Gutti, R.K. Wnt Signaling: Role in Regulation of Haematopoiesis. Indian J Hematol Blood Transfus. 2016, 32, 123–134. [Google Scholar] [CrossRef]
  48. Soares-Lima, S.C.; Pombo-de-Oliveira, M.S.; Carneiro, F.R.G. The multiple ways Wnt signaling contributes to acute leukemia pathogenesis. J Leukoc Biol. 2020, 108, 1081–1099. [Google Scholar] [CrossRef]
  49. Yan, K.; Tian, J.; Shi, W.; Xia, H.; Zhu, Y. LncRNA SNHG6 is Associated with Poor Prognosis of Gastric Cancer and Promotes Cell Proliferation and EMT through Epigenetically Silencing p27 and Sponging miR-101-3p. Cell Physiol Biochem. 2017, 42, 999–1012. [Google Scholar] [CrossRef]
  50. Wu, X.; Zhou, J.; Wu, Z.; Chen, C.; Liu, J.; Wu, G.; Zhai, J.; Liu, F.; Li, G. miR-101-3p Suppresses HOX Transcript Antisense RNA (HOTAIR)-Induced Proliferation and Invasion Through Directly Targeting SRF in Gastric Carcinoma Cells. Oncol Res. 2017, 25, 1383–1390. [Google Scholar] [CrossRef]
  51. Motofei, I.G. Biology of cancer; from cellular and molecular mechanisms to developmental processes and adaptation. Semin Cancer Biol. 2021, 23, S1044. [Google Scholar] [CrossRef]
  52. Bao, R.F.; Shu, Y.J.; Hu, Y.P.; Wang, X.A.; Zhang, F.; Liang, H.B.; Ye, Y.Y.; Li, H.F.; Xiang, S.S.; Weng, H.; Cao, Y.; Wu, X.S.; Li, M.L.; Wu, W.G.; Zhang, Y.J.; Jiang, L.; Dong, Q.; Liu, Y.B. miR-101 targeting ZFX suppresses tumor proliferation and metastasis by regulating the MAPK/Erk and Smad pathways in gallbladder carcinoma. Oncotarget. 2016, 7, 22339–22354. [Google Scholar] [CrossRef] [PubMed]
  53. Huang, Y.; Zou, Y.; Lin, L.; Ma, X.; Zheng, R. miR-101 regulates cell proliferation and apoptosis by targeting KDM1A in diffuse large B cell lymphoma. Cancer Manag Res. 2019, 11, 2739–2746. [Google Scholar] [CrossRef] [PubMed]
  54. Parascandolo, A.; Laukkanen, M.O. Carcinogenesis and Reactive Oxygen Species Signaling: Interaction of the NADPH Oxidase NOX1-5 and Superoxide Dismutase 1-3 Signal Transduction Pathways. Antioxid Redox Signal. 2019, 30, 443–486. [Google Scholar] [CrossRef]
  55. Lang, J.Y.; Ma, K.; Guo, J.X.; Sun, H. Oxidative stress induces B lymphocyte DNA damage and apoptosis by upregulating p66shc. Eur Rev Med Pharmacol Sci. 2018, 22, 1051–1060. [Google Scholar] [CrossRef]
  56. Zhou, F.; Shen, Q.; Claret, F.X. Novel roles of reactive oxygen species in the pathogenesis of acute myeloid leukemia. J Leukoc Biol. 2013, 94, 423–429. [Google Scholar] [CrossRef]
  57. Wang, L.; Howell, M.E.A.; Sparks-Wallace, A.; Hawkins, C.; Nicksic, C.A.; Kohne, C.; Hall, K.H.; Moorman, J.P.; Yao, Z.Q.; Ning, S. p62-mediated Selective autophagy endows virus-transformed cells with insusceptibility to DNA damage under oxidative stress. PLoS Pathog. 2019, 15, e1007541. [Google Scholar] [CrossRef]
  58. Moniczewski, A.; Gawlik, M.; Smaga, I.; Niedzielska, E.; Krzek, J.; Przegaliński, E.; Pera, J.; Filip, M. Oxidative stress as an etiological factor and a potential treatment target of psychiatric disorders. Part 1. Chemical aspects and biological sources of oxidative stress in the brain. Pharmacol Rep. 2015, 67, 560–568. [Google Scholar] [CrossRef]
  59. Liguori, I.; Russo, G.; Curcio, F.; Bulli, G.; Aran, L.; Della-Morte, D.; Gargiulo, G.; Testa, G.; Cacciatore, F.; Bonaduce, D.; Abete, P. Oxidative stress, aging, and diseases. Clin Interv Aging. 2018, 13, 757–772. [Google Scholar] [CrossRef]
  60. Ma, Y.; Chapman, J.; Levine, M.; Polireddy, K.; Drisko, J.; Chen, Q. High-dose parenteral ascorbate enhanced chemosensitivity of ovarian cancer and reduced toxicity of chemotherapy. Sci Transl Med. 2014, 6, 222ra18. [Google Scholar] [CrossRef]
  61. Wu, X.; Cheng, J.; Wang, X. Dietary Antioxidants: Potential Anticancer Agents. Nutr Cancer. 2017, 69, 521–533. [Google Scholar] [CrossRef]
  62. Breitenbach, M.; Eckl, P. Introduction to Oxidative Stress in Biomedical and Biological Research. Biomolecules. 2015, 5, 1169–1177. [Google Scholar] [CrossRef] [PubMed]
  63. Simoncini, C.; Orsucci, D.; Caldarazzo Ienco, E.; Siciliano, G.; Bonuccelli, U.; Mancuso, M. Alzheimer’s pathogenesis and its link to the mitochondrion. Oxid Med Cell Longev. 2015, 2015, 803942. [Google Scholar] [CrossRef] [PubMed]
  64. Smaga, I.; Niedzielska, E.; Gawlik, M.; Moniczewski, A.; Krzek, J.; Przegaliński, E.; Pera, J.; Filip, M. Oxidative stress as an etiological factor and a potential treatment target of psychiatric disorders. Part 2. Depression, anxiety, schizophrenia and autism. Pharmacol Rep. 2015, 67, 569–580. [Google Scholar] [CrossRef] [PubMed]
  65. Georgescu, T.A.; Bohiltea, R.; Munteanu, O.; Grigoriu, C.; Paunica, I.; Sajin, M. A mini-review regarding the carcinogenesis and morphology of serous tumors of the ovary, fallopian tube and peritoneum. J Mind Med Sci. 2021, 8, 44–52. [Google Scholar] [CrossRef]
  66. Tucker, P.S.; Scanlan, A.T.; Dalbo, V.J. Chronic kidney disease influences multiple systems: describing the relationship between oxidative stress, inflammation, kidney damage, and concomitant disease. Oxid Med Cell Longev. 2015, 2015, 806358. [Google Scholar] [CrossRef]
  67. Kleinstern, G.; Maurer, M.J.; Liebow, M.; Habermann, T.M.; Koff, J.L.; Allmer, C.; Witzig, T.E.; Nowakowski, G.S.; Micallef, I.N.; Johnston, P.B.; Inwards, D.J.; Thompson, C.A.; Feldman, A.L.; Link, B.K.; Flowers, C.; Slager, S.L.; Cerhan, J.R. History of autoimmune conditions and lymphoma prognosis. Blood Cancer, J. 2018, 8, 73. [Google Scholar] [CrossRef]
  68. Yang, S.; Li, J.Y.; Xu, W. Role of BAFF/BAFF-R axis in B-cell non-Hodgkin lymphoma. Crit Rev Oncol Hematol. 2014, 91, 113–122. [Google Scholar] [CrossRef]
  69. Liu, K.; Liu, X.; Wang, M.; Wang, X.; Kang, H.; Lin, S.; Yang, P.; Dai, C.; Xu, P.; Li, S.; Dai, Z. Two common functional catalase gene polymorphisms (rs1001179 and rs794316) and cancer susceptibility: evidence from 14,942 cancer cases and 43,285 controls. Oncotarget. 2016, 7, 62954–62965. [Google Scholar] [CrossRef]
  70. Makgoeng, S.B.; Bolanos, R.S.; Jeon, C.Y.; Weiss, R.E.; Arah, O.A.; Breen, E.C.; Martínez-Maza, O.; Hussain, S.K. Markers of Immune Activation and Inflammation, and Non-Hodgkin Lymphoma: A Meta-Analysis of Prospective Studies. JNCI Cancer Spectr. 2018, 2, pky082. [Google Scholar] [CrossRef]
  71. Gerondakis, S.; Siebenlist, U. Roles of the NF-kappaB pathway in lymphocyte development and function. Cold Spring Harb Perspect Biol. 2010, 2, a000182. [Google Scholar] [CrossRef]
  72. Gaman, M.A.; Epingeac, M.E.; Gaman, A.M. Evaluation of oxidative stress and high-density lipoprotein cholesterol levels in diffuse large B-cell lymphoma. Rev Chim. 2019, 70, 977–980. [Google Scholar] [CrossRef]
  73. Wang, S.S.; Davis, S.; Cerhan, J.R.; Hartge, P.; Severson, R.K.; Cozen, W.; Lan, Q.; Welch, R.; Chanock, S.J.; Rothman, N. Polymorphisms in oxidative stress genes and risk for non-Hodgkin lymphoma. Carcinogenesis. 2006, 27, 1828–1834. [Google Scholar] [CrossRef] [PubMed]
  74. Kim, H.N.; Kim, N.Y.; Yu, L.; Kim, Y.K.; Lee, I.K.; Yang, D.H.; Lee, J.J.; Shin, M.H.; Park, K.S.; Choi, J.S.; Kim, H.J. Polymorphisms in DNA repair genes and MDR1 and the risk for non-Hodgkin lymphoma. Int J Mol Sci. 2014, 15, 6703–6716. [Google Scholar] [CrossRef] [PubMed]
  75. Thanan, R.; Oikawa, S.; Hiraku, Y.; Ohnishi, S.; Ma, N.; Pinlaor, S.; Yongvanit, P.; Kawanishi, S.; Murata, M. Oxidative stress and its significant roles in neurodegenerative diseases and cancer. Int J Mol Sci. 2014, 16, 193–217. [Google Scholar] [CrossRef]
  76. Kawanishi, S.; Ohnishi, S.; Ma, N.; Hiraku, Y.; Murata, M. Crosstalk between DNA Damage and Inflammation in the Multiple Steps of Carcinogenesis. Int J Mol Sci. 2017, 18, 1808. [Google Scholar] [CrossRef]
  77. Benzer, F.; Kandemir, F.M.; Ozkaraca, M.; Kucukler, S.; Caglayan, C. Curcumin ameliorates doxorubicin-induced cardiotoxicity by abrogation of inflammation, apoptosis, oxidative DNA damage, and protein oxidation in rats. J Biochem Mol Toxicol. 2018, 32. [Google Scholar] [CrossRef]
  78. Mumcu, U.Y.; Kocer, I.; Ates, O.; Alp, H.H. Decreased paraoxonase1 activity and increased malondialdehyde and oxidative DNA damage levels in primary open angle glaucoma. Int J Ophthalmol. 2016, 9, 1518–1520. [Google Scholar] [CrossRef]
  79. Black, C.N.; Bot, M.; Révész, D.; Scheffer, P.G.; Penninx, B. The association between three major physiological stress systems and oxidative DNA and lipid damage. Psychoneuroendocrinology. 2017, 80, 56–66. [Google Scholar] [CrossRef]
  80. El-Mezayen, H.A.; Darwish, H.; Hasheim, M.; El-Baz, H.A. Oxidant/Antioxidant Status and their Relations to Chemotherapy in Non-Hodgkin’s Lymphoma. International Journal of Pharmaceutical and Clinical Research. 2015, 7, 269–274. [Google Scholar]
  81. Jakovcevic, D.; Dedic-Plavetic, N.; Vrbanec, D.; Jakovcevic, A.; Jakic-Razumovic, J. Breast Cancer Molecular Subtypes and Oxidative DNA Damage. Appl Immunohistochem Mol Morphol. 2015, 23, 696–703. [Google Scholar] [CrossRef]
  82. Ma-On, C.; Sanpavat, A.; Whongsiri, P.; Suwannasin, S.; Hirankarn, N.; Tangkijvanich, P.; Boonla, C. Oxidative stress indicated by elevated expression of Nrf2 and 8-OHdG promotes hepatocellular carcinoma progression. Med Oncol. 2017, 34, 57. [Google Scholar] [CrossRef] [PubMed]
  83. He, H.; Zhao, Y.; Wang, N.; Zhang, L.; Wang, C. 8-Hydroxy-2’-deoxyguanosine expression predicts outcome of esophageal cancer. Ann Diagn Pathol. 2014, 18, 326–328. [Google Scholar] [CrossRef] [PubMed]
  84. Kale, I. The predictive role of monocyte-lymphocyte ratio and platelet-lymphocyte ratio in postmenopausal osteoporosis. J Clin Invest Surg. 2021, 6, 141–147. [Google Scholar] [CrossRef]
  85. Fenga, C.; Gangemi, S.; Costa, C. Benzene exposure is associated with epigenetic changes (Review). Mol Med Rep. 2016, 13, 3401–3405. [Google Scholar] [CrossRef]
  86. Arakaki, H.; Osada, Y.; Takanashi, S.; Ito, C.; Aisa, Y.; Nakazato, T. Oxidative Stress Is Associated with Poor Prognosis in Patients with Follicular Lymphoma. Blood. 2016, 128, 1787. [Google Scholar] [CrossRef]
  87. Fenga, C.; Gangemi, S.; Teodoro, M.; Rapisarda, V.; Golokhvast, K.; Docea, A.O.; Tsatsakis, A.M.; Costa, C. 8-Hydroxydeoxyguanosine as a biomarker of oxidative DNA damage in workers exposed to low-dose benzene. Toxicol Rep. 2017, 4, 291–295. [Google Scholar] [CrossRef]
  88. Kantner, H.P.; Warsch, W.; Delogu, A.; Bauer, E.; Esterbauer, H.; Casanova, E.; Sexl, V.; Stoiber, D. ETV6/RUNX1 induces reactive oxygen species and drives the accumulation of DNA damage in B cells. Neoplasia. 2013, 15, 1292–1300. [Google Scholar] [CrossRef]
  89. Rochette, L.; Guenancia, C.; Gudjoncik, A.; Hachet, O.; Zeller, M.; Cottin, Y.; Vergely, C. Anthracyclines/ trastuzumab: new aspects of cardiotoxicity and molecular mechanisms. Trends Pharmacol Sci. 2015, 36, 326–348. [Google Scholar] [CrossRef]
  90. Geybels, M.S.; van den Brandt, P.A.; van Schooten, F.J.; Verhage, B.A. Oxidative stress-related genetic variants, pro-and antioxidant intake and status, and advanced prostate cancer risk. Cancer Epidemiol Biomarkers Prev. 2015, 24, 178–186. [Google Scholar] [CrossRef]
  91. Wang, C.; Zhang, R.; Chen, N.; Yang, L.; Wang, Y.; Sun, Y.; Huang, L.; Zhu, M.; Ji, Y.; Li, W. Association between glutathione peroxidase-1 (GPX1) Rs1050450 polymorphisms and cancer risk. Int J Clin Exp Pathol. 2017, 10, 9527–9540. [Google Scholar]
  92. Tasaki, E.; Sakurai, H.; Nitao, M.; Matsuura, K.; Iuchi, Y. Uric acid, an important antioxidant contributing to survival in termites. PLoS One. 2017, 12, e0179426. [Google Scholar] [CrossRef]
  93. Liu, D.; Yun, Y.; Yang, D.; Hu, X.; Dong, X.; Zhang, N.; Zhang, L.; Yin, H.; Duan, W. What Is the Biological Function of Uric Acid? An Antioxidant for Neural Protection or a Biomarker for Cell Death. Dis Markers. 2019, 2019, 4081962. [Google Scholar] [CrossRef] [PubMed]
  94. Yasutake, Y.; Tomita, K.; Higashiyama, M.; Furuhashi, H.; Shirakabe, K.; Takajo, T.; Maruta, K.; Sato, H.; Narimatsu, K.; Yoshikawa, K.; Okada, Y.; Kurihara, C.; Watanabe, C.; Komoto, S.; Nagao, S.; Matsuo, H.; Miura, S.; Hokari, R. Uric acid ameliorates indomethacin-induced enteropathy in mice through its antioxidant activity. J Gastroenterol Hepatol. 2017, 32, 1839–1845. [Google Scholar] [CrossRef]
  95. Itahana, Y.; Han, R.; Barbier, S.; Lei, Z.; Rozen, S.; Itahana, K. The uric acid transporter SLC2A9 is a direct target gene of the tumor suppressor p53 contributing to antioxidant defense. Oncogene. 2015, 34, 1799–1810. [Google Scholar] [CrossRef]
  96. Trzeciecka, A.; Klossowski, S.; Bajor, M.; Zagozdzon, R.; Gaj, P.; Muchowicz, A.; Malinowska, A.; Czerwoniec, A.; Barankiewicz, J.; Domagala, A.; Chlebowska, J.; Prochorec-Sobieszek, M.; Winiarska, M.; Ostaszewski, R.; Gwizdalska, I.; Golab, J.; Nowis, D.; Firczuk, M. Dimeric peroxiredoxins are druggable targets in human Burkitt lymphoma. Oncotarget. 2016, 7, 1717–1731. [Google Scholar] [CrossRef]
  97. Muchowicz, A.; Firczuk, M.; Chlebowska, J.; Nowis, D.; Stachura, J.; Barankiewicz, J.; Trzeciecka, A.; Kłossowski, S.; Ostaszewski, R.; Zagożdżon, R.; Pu, J.X.; Sun, H.D.; Golab, J. Adenanthin targets proteins involved in the regulation of disulphide bonds. Biochem Pharmacol. 2014, 89, 210–216. [Google Scholar] [CrossRef]
  98. Fiskus, W.; Saba, N.; Shen, M.; Ghias, M.; Liu, J.; Gupta, S.D.; Chauhan, L.; Rao, R.; Gunewardena, S.; Schorno, K.; Austin, C.P.; Maddocks, K.; Byrd, J.; Melnick, A.; Huang, P.; Wiestner, A.; Bhalla, K.N. Auranofin induces lethal oxidative and endoplasmic reticulum stress and exerts potent preclinical activity against chronic lymphocytic leukemia. Cancer Res. 2014, 74, 2520–2532. [Google Scholar] [CrossRef]
  99. Celegato, M.; Borghese, C.; Casagrande, N.; Mongiat, M.; Kahle, X.U.; Paulitti, A.; Spina, M.; Colombatti, A.; Aldinucci, D. Preclinical activity of the repurposed drug auranofin in classical Hodgkin lymphoma. Blood. 2015, 126, 1394–1397. [Google Scholar] [CrossRef]
  100. Bansal, A.; Simon, M.C. Glutathione metabolism in cancer progression and treatment resistance. J Cell Biol. 2018, 217, 2291–2298. [Google Scholar] [CrossRef]
  101. Kiebala, M.; Skalska, J.; Casulo, C.; Brookes, P.S.; Peterson, D.R.; Hilchey, S.P.; Dai, Y.; Grant, S.; Maggirwar, S.B.; Bernstein, S.H. Dual targeting of the thioredoxin and glutathione antioxidant systems in malignant B cells: a novel synergistic therapeutic approach. Exp Hematol. 2015, 43, 89–99. [Google Scholar] [CrossRef]
  102. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2019. CA Cancer J Clin. 2019, 69, 7–34. [Google Scholar] [CrossRef] [PubMed]
  103. Scott, D.W.; King, R.L.; Staiger, A.M.; Ben-Neriah, S.; Jiang, A.; Horn, H.; Mottok, A.; Farinha, P.; Slack, G.W.; Ennishi, D.; Schmitz, N.; Pfreundschuh, M.; Nowakowski, G.S.; Kahl, B.S.; Connors, J.M.; Gascoyne, R.D.; Ott, G.; Macon, W.R.; Rosenwald, A. High-grade B-cell lymphoma with MYC and BCL2 and/or BCL6 rearrangements with diffuse large B-cell lymphoma morphology. Blood. 2018, 131, 2060–2064. [Google Scholar] [CrossRef] [PubMed]
  104. 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.; Hodson, D.J.; Xiao, W.; Yu, X.; Yang, Y.; Zhao, H.; Xu, W.; Liu, X.; Zhou, B.; Du, W.; Chan, W.C.; Jaffe, E.S.; Gascoyne, R.D.; Connors, J.M.; Campo, E.; Lopez-Guillermo, A.; Rosenwald, A.; Ott, G.; Delabie, J.; Rimsza, L.M.; Tay Kuang Wei, K.; Zelenetz, A.D.; Leonard, J.P.; Bartlett, N.L.; Tran, B.; Shetty, J.; Zhao, Y.; Soppet, D.R.; Pittaluga, S.; Wilson, W.H.; Staudt, L.M. Genetics and Pathogenesis of Diffuse Large B-Cell Lymphoma. N Engl J Med. 2018, 378, 1396–1407. [Google Scholar] [CrossRef]
  105. Kinowaki, Y.; Kurata, M.; Ishibashi, S.; Ikeda, M.; Tatsuzawa, A.; Yamamoto, M.; Miura, O.; Kitagawa, M.; Yamamoto, K. Glutathione peroxidase 4 overexpression inhibits ROS-induced cell death in diffuse large B-cell lymphoma. Lab Invest. 2018, 98, 609–619. [Google Scholar] [CrossRef]
  106. Zhang, J.; Li, X.; Han, X.; Liu, R.; Fang, J. Targeting the Thioredoxin System for Cancer Therapy. Trends Pharmacol Sci. 2017, 38, 794–808. [Google Scholar] [CrossRef]
  107. Jia, J.J.; Geng, W.S.; Wang, Z.Q.; Chen, L.; Zeng, X.S. The role of thioredoxin system in cancer: strategy for cancer therapy. Cancer Chemother Pharmacol. 2019, 84, 453–470. [Google Scholar] [CrossRef]
  108. Wang, S.; Lu, Y.; Woods, K.; Di Trapani, G.; Tonissen, K.F. Investigating the Thioredoxin and Glutathione Systems’ Response in Lymphoma Cells after Treatment with [Au(d2pype)2]CL. Antioxidants (Basel). 2021, 10, 104. [Google Scholar] [CrossRef]
  109. Sewastianik, T.; Szydlowski, M.; Jablonska, E.; Bialopiotrowicz, E.; Kiliszek, P.; Gorniak, P.; Polak, A.; Prochorec-Sobieszek, M.; Szumera-Cieckiewicz, A.; Kaminski, T.S.; Markowicz, S.; Nowak, E.; Grygorowicz, M.A.; Warzocha, K.; Juszczynski, P. FOXO1 is a TXN-and p300-dependent sensor and effector of oxidative stress in diffuse large B-cell lymphomas characterized by increased oxidative metabolism. Oncogene. 2016, 35, 5989–6000. [Google Scholar] [CrossRef]
  110. Chan, M.C.; Savela, J.; Ollikainen, R.K.; Teppo, H.R.; Miinalainen, I.; Pirinen, R.; Kari, E.J.M.; Kuitunen, H.; Turpeenniemi-Hujanen, T.; Kuittinen, O.; Kuusisto, M.E.L. Testis-Specific Thioredoxins TXNDC2, TXNDC3, and TXNDC6 Are Expressed in Both Testicular and Systemic DLBCL and Correlate with Clinical Disease Presentation. Oxid Med Cell Longev. 2021, 2021, 8026941. [Google Scholar] [CrossRef]
  111. Mai, Y.; Yu, J.J.; Bartholdy, B.; Xu-Monette, Z.Y.; Knapp, E.E.; Yuan, F.; Chen, H.; Ding, B.B.; Yao, Z.; Das, B.; Zou, Y.; Young, K.H.; Parekh, S.; Ye, B.H. An oxidative stress-based mechanism of doxorubicin cytotoxicity suggests new therapeutic strategies in ABC-DLBCL. Blood. 2016, 128, 2797–2807. [Google Scholar] [CrossRef]
  112. Freedman, A. Follicular lymphoma: 2018 update on diagnosis and management. Am J Hematol. 2018, 93, 296–305. [Google Scholar] [CrossRef] [PubMed]
  113. Louissaint, A., Jr.; Schafernak, K.T.; Geyer, J.T.; Kovach, A.E.; Ghandi, M.; Gratzinger, D.; Roth, C.G.; Paxton, C.N.; Kim, S.; Namgyal, C.; Morin, R.; Morgan, E.A.; Neuberg, D.S.; South, S.T.; Harris, M.H.; Hasserjian, R.P.; Hochberg, E.P.; Garraway, L.A.; Harris, N.L.; Weinstock, D.M. Pediatric-type nodal follicular lymphoma: a biologically distinct lymphoma with frequent MAPK pathway mutations. Blood. 2016, 128, 1093–1100. [Google Scholar] [CrossRef] [PubMed]
  114. Peroja, P.; Haapasaari, K.M.; Mannisto, S.; Miinalainen, I.; Koivunen, P.; Leppä, S.; Karjalainen-Lindsberg, M.L.; Kuusisto, M.E.; Turpeenniemi-Hujanen, T.; Kuittinen, O.; Karihtala, P. Total peroxiredoxin expression is associated with survival in patients with follicular lymphoma. Virchows Arch. 2016, 468, 623–630. [Google Scholar] [CrossRef]
  115. Pieters, T.; T’Sas, S.; Vanhee, S.; Almeida, A.; Driege, Y.; Roels, J.; Van Loocke, W.; Daneels, W.; Baens, M.; Marchand, A.; Van Trimpont, M.; Matthijssens, F.; Morscio, J.; Lemeire, K.; Lintermans, B.; Reunes, L.; Chaltin, P.; Offner, F.; Van Dorpe, J.; Hochepied, T.; Berx, G.; Beyaert, R.; Staal, J.; Van Vlierberghe, P.; Goossens, S. Cyclin D2 overexpression drives B1a-derived MCL-like lymphoma in mice. J Exp Med. 2021, 218, e20202280. [Google Scholar] [CrossRef]
  116. Zhang, H.; Chen, Z.; Miranda, R.N.; Medeiros, L.J.; McCarty, N. Bifurcated BACH2 control coordinates mantle cell lymphoma survival and dispersal during hypoxia. Blood. 2017, 130, 763–776. [Google Scholar] [CrossRef]
  117. Tafani, M.; Sansone, L.; Limana, F.; Arcangeli, T.; De Santis, E.; Polese, M.; Fini, M.; Russo, M.A. The Interplay of Reactive Oxygen Species, Hypoxia, Inflammation, and Sirtuins in Cancer Initiation and Progression. Oxid Med Cell Longev. 2016, 2016, 3907147. [Google Scholar] [CrossRef]
  118. Marcelis, L.; Tousseyn, T.; Sagaert, X. MALT Lymphoma as a Model of Chronic Inflammation-Induced Gastric Tumor Development. Curr Top Microbiol Immunol. 2019, 421, 77–106. [Google Scholar] [CrossRef]
  119. Violeta Filip, P.; Cuciureanu, D.; Sorina Diaconu, L.; Maria Vladareanu, A.; Silvia Pop, C. MALT lymphoma: epidemiology, clinical diagnosis and treatment. J Med Life. 2018, 11, 187–193. [Google Scholar] [CrossRef]
  120. Juárez-Salcedo, L.M.; Sokol, L.; Chavez, J.C.; Dalia, S. Primary Gastric Lymphoma, Epidemiology, Clinical Diagnosis, and Treatment. Cancer Control. 2018, 25, 1073274818778256. [Google Scholar] [CrossRef]
  121. Delgado, J.; Nadeu, F.; Colomer, D.; Campo, E. Chronic lymphocytic leukemia: from molecular pathogenesis to novel therapeutic strategies. Haematologica. 2020, 105, 2205–2217. [Google Scholar] [CrossRef]
  122. Baliakas, P.; Jeromin, S.; Iskas, M.; Puiggros, A.; Plevova, K.; Nguyen-Khac, F.; Davis, Z.; Rigolin, G.M.; Visentin, A.; Xochelli, A.; Delgado, J.; Baran-Marszak, F.; Stalika, E.; Abrisqueta, P.; Durechova, K.; Papaioannou, G.; Eclache, V.; Dimou, M.; Iliakis, T.; Collado, R.; Doubek, M.; Calasanz, M.J.; Ruiz-Xiville, N.; Moreno, C.; Jarosova, M.; Leeksma, A.C.; Panayiotidis, P.; Podgornik, H.; Cymbalista, F.; Anagnostopoulos, A.; Trentin, L.; Stavroyianni, N.; Davi, F.; Ghia, P.; Kater, A.P.; Cuneo, A.; Pospisilova, S.; Espinet, B.; Athanasiadou, A.; Oscier, D.; Haferlach, C.; Stamatopoulos, K.; ERIC, the European Research Initiative on CLL. Cytogenetic complexity in chronic lymphocytic leukemia: definitions, associations, and clinical impact. Blood. 2019, 133, 1205–1216. [Google Scholar] [CrossRef] [PubMed]
  123. Sabry, S.A.; El-Senduny, F.; Abousamra, N.; El-Din, M.S. Interaction between Th9, interleukin-9 and oxidative stress in chronic lymphocytic leukemia. Trends in Applied Sciences Research. 2019, 14, 56–69. [Google Scholar] [CrossRef]
  124. Zhevak, T.; Shelekhova, T.; Chesnokova, N.; Tsareva, O.; Chanturidze, A.; Litvitsky, P.; Andriutsa, N.; Samburova, N.; Budnik, I. The relationship between oxidative stress and cytogenetic abnormalities in B-cell chronic lymphocytic leukemia. Exp Mol Pathol. 2020, 116, 104524. [Google Scholar] [CrossRef]
  125. Sabry, S.A.; El-Senduny, F.F.; Abousamra, N.K.; Salah El-Din, M.; Youssef, M.M. Oxidative stress in CLL patients leads to activation of Th9 cells: an experimental and comprehensive survey. Immunol Med. 2020, 43, 36–46. [Google Scholar] [CrossRef]
  126. Zhang, J.; Lei, W.; Chen, X.; Wang, S.; Qian, W. Oxidative stress response induced by chemotherapy in leukemia treatment. Mol Clin Oncol. 2018, 8, 391–399. [Google Scholar] [CrossRef]
  127. Moignet, A.; Hasanali, Z.; Zambello, R.; Pavan, L.; Bareau, B.; Tournilhac, O.; Roussel, M.; Fest, T.; Awwad, A.; Baab, K.; Semenzato, G.; Houot, R.; Loughran, T.P., Jr.; Lamy, T. Cyclophosphamide as a first-line therapy in LGL leukemia. Leukemia. 2014, 28, 1134–1136. [Google Scholar] [CrossRef]
  128. Alqahtani, S.; Mahmoud, A.M. Gamma-Glutamylcysteine Ethyl Ester Protects against Cyclophosphamide-Induced Liver Injury and Hematologic Alterations via Upregulation of PPARγ and Attenuation of Oxidative Stress, Inflammation, and Apoptosis. Oxid Med Cell Longev. 2016, 2016, 4016209. [Google Scholar] [CrossRef]
  129. Mohammad, M.K.; Avila, D.; Zhang, J.; Barve, S.; Arteel, G.; McClain, C.; Joshi-Barve, S. Acrolein cytotoxicity in hepatocytes involves endoplasmic reticulum stress, mitochondrial dysfunction and oxidative stress. Toxicol Appl Pharmacol. 2012, 265, 73–82. [Google Scholar] [CrossRef]
  130. McGowan, J.V.; Chung, R.; Maulik, A.; Piotrowska, I.; Walker, J.M.; Yellon, D.M. Anthracycline Chemotherapy and Cardiotoxicity. Cardiovasc Drugs Ther. 2017, 31, 63–75. [Google Scholar] [CrossRef]
  131. Teppo, H.R.; Soini, Y.; Karihtala, P. Reactive Oxygen Species-Mediated Mechanisms of Action of Targeted Cancer Therapy. Oxid Med Cell Longev. 2017, 2017, 1485283. [Google Scholar] [CrossRef]
  132. Spiers, J.G.; Chen, H.J.; Sernia, C.; Lavidis, N.A. Activation of the hypothalamic-pituitary-adrenal stress axis induces cellular oxidative stress. Front Neurosci. 2015, 8, 456. [Google Scholar] [CrossRef] [PubMed]
  133. Hubenak, J.R.; Zhang, Q.; Branch, C.D.; Kronowitz, S.J. Mechanisms of injury to normal tissue after radiotherapy: a review. Plast Reconstr Surg. 2014, 133, 49e–56e. [Google Scholar] [CrossRef] [PubMed]
  134. Castaldo, S.A.; Freitas, J.R.; Conchinha, N.V.; Madureira, P.A. The Tumorigenic Roles of the Cellular REDOX Regulatory Systems. Oxid Med Cell Longev. 2016, 2016, 8413032. [Google Scholar] [CrossRef]
  135. ALHaithloul, H.A.S.; Alotaibi, M.F.; Bin-Jumah, M.; Elgebaly, H.; Mahmoud, A.M. Olea europaea leaf extract up-regulates Nrf2/ARE/HO-1 signaling and attenuates cyclophosphamide-induced oxidative stress, inflammation and apoptosis in rat kidney. Biomed Pharmacother. 2019, 111, 676–685. [Google Scholar] [CrossRef]
  136. Yardim, A.; Kandemir, F.M.; Ozdemir, S.; Kucukler, S.; Comakli, S.; Gur, C.; Celik, H. Quercetin provides protection against the peripheral nerve damage caused by vincristine in rats by suppressing caspase 3, NF-κB, ATF-6 pathways and activating Nrf2, Akt pathways. Neurotoxicology. 2020, 81, 137–146. [Google Scholar] [CrossRef]
  137. Găman, A.M.; Egbuna, C.; Găman, M.A. Natural bioactive lead compounds effective against haematological malignancies. In Egbuna, C.; Kumar, S., Ifemeje, J.C., Ezzat, S.M., Kaliyaperumal, S. (Eds.). Phytochemicals as Lead Compounds for New Drug Discovery, Eds.; Elsevier: Amsterdam, The Netherlands, 2020; pp. 95–115. [Google Scholar] [CrossRef]
  138. Zhang, Y.; Jiang, X.; Li, X.; Găman, M.A.; Kord-Varkaneh, H.; Rahmani, J.; Salehi-Sahlabadi, A.; Day, A.S.; Xu, Y. Serum Vitamin D Levels and Risk of Liver Cancer: A Systematic Review and Dose-Response Meta-Analysis of Cohort Studies. Nutr Cancer. 2021, 73, 1–9. [Google Scholar] [CrossRef]
  139. Tofolean, I.T.; Ganea, C.; Ionescu, D.; Filippi, A.; Garaiman, A.; Goicea, A.; Gaman, M.A.; Dimancea, A.; Baran, I. Cellular determinants involving mitochondrial dysfunction, oxidative stress and apoptosis correlate with the synergic cytotoxicity of epigallocatechin-3-gallate and menadione in human leukemia Jurkat T cells. Pharmacol Res. 2016, 103, 300–17. [Google Scholar] [CrossRef]
  140. Motofei, I.G. Nobel Prize for immune checkpoint inhibitors, understanding the immunological switching between immunosuppression and autoimmunity. Expert Opin Drug Saf. 2021, 1–14. [Google Scholar] [CrossRef]
  141. Montazeri, R.S.; Fatahi, S.; Sohouli, M.H.; Abu-Zaid, A.; Santos, H.O.; Găman, M.A.; Shidfar, F. The effect of nigella sativa on biomarkers of inflammation and oxidative stress: A systematic review and meta-analysis of randomized controlled trials. J Food Biochem. 2021, 45, e13625. [Google Scholar] [CrossRef]
  142. Sánchez, A.; Calpena, A.C.; Clares, B. Evaluating the Oxidative Stress in Inflammation: Role of Melatonin. Int J Mol Sci. 2015, 16, 16981–17004. [Google Scholar] [CrossRef]
  143. 143Liu, J.; Clough, S.J.; Hutchinson, A.J.; Adamah-Biassi, E.B.; Popovska-Gorevski, M.; Dubocovich, M.L. MT1 and MT2 Melatonin Receptors: A Therapeutic Perspective. Annu Rev Pharmacol Toxicol. 2016, 56, 361–83. [Google Scholar] [CrossRef]
  144. Wang, T.H.; Hsueh, C.; Chen, C.C.; Li, W.S.; Yeh, C.T.; Lian, J.H.; Chang, J.L.; Chen, C.Y. Melatonin Inhibits the Progression of Hepatocellular Carcinoma through MicroRNA Let7i-3p Mediated RAF1 Reduction. Int J Mol Sci. 2018, 19, 2687. [Google Scholar] [CrossRef]
  145. Sánchez, D.I.; González-Fernández, B.; Crespo, I.; San-Miguel, B.; Álvarez, M.; González-Gallego, J.; Tuñón, M.J. Melatonin modulates dysregulated circadian clocks in mice with diethylnitrosamine-induced hepatocellular carcinoma. J Pineal Res. 2018, 65, e12506. [Google Scholar] [CrossRef]
  146. Yan, G.; Lei, H.; He, M.; Gong, R.; Wang, Y.; He, X.; Li, G.; Pang, P.; Li, X.; Yu, S.; Du, W.; Yuan, Y. Melatonin triggers autophagic cell death by regulating RORC in Hodgkin lymphoma. Biomed Pharmacother. 2020, 123, 109811. [Google Scholar] [CrossRef]
  147. Li, Y.; Li, S.; Zhou, Y.; Meng, X.; Zhang, J.J.; Xu, D.P.; Li, H.B. Melatonin for the prevention and treatment of cancer. Oncotarget. 2017, 8, 39896–39921. [Google Scholar] [CrossRef]
  148. Mehrzadi, M.H.; Hosseinzadeh, A.; Juybari, K.B.; Mehrzadi, S. Melatonin and urological cancers: a new therapeutic approach. Cancer Cell Int. 2020, 20, 444. [Google Scholar] [CrossRef]
  149. Gaman, A.M.; Buga, A.M.; Gaman, M.A.; Popa-Wagner, A. The role of oxidative stress and the effects of antioxidants on the incidence of infectious complications of chronic lymphocytic leukemia. Oxid Med Cell Longev. 2014, 2014, 158135. [Google Scholar] [CrossRef]
  150. Gaman, A.M.; Moisa, C.; Diaconu, C.C.; Gaman, M.A. Crosstalk between Oxidative Stress, Chronic Inflammation and Disease Progression in Essential Thrombocythemia. Rev. Chim. 2019, 70, 3486–3489. [Google Scholar] [CrossRef]
  151. Moisa, C.; Gaman, M.A.; Diaconu, C.C.; Gaman, A.M. Oxidative Stress Levels, JAK2V617F Mutational Status and Thrombotic Complications in Patients with Essential Thrombocythemia. Rev. Chim. 2019, 70, 2822–2825. [Google Scholar] [CrossRef]
  152. Găman, M.A.; Cozma, M.A.; Dobrică, E.C.; Crețoiu, S.M.; Găman, A.M.; Diaconu, C.C. Liquid Biopsy and Potential Liquid Biopsy-Based Biomarkers in Philadelphia- Negative Classical Myeloproliferative Neoplasms: A Systematic Review. Life (Basel) 2021, 11, 677. [Google Scholar] [CrossRef]
Table 1. Risk factors for NHL.
Table 1. Risk factors for NHL.
Viral infections
Epstein-Barr virus is related with the endemic variant of BL [11];
HTLV-1 causes adult T-cell lymphomas [12];
Human Herpes virus-8 is associated with Kaposi sarcoma [13];
Hepatitis B and C -DLBCL and splenic marginal zone lymphoma are the most frequent subtypes due to the infection with the hepatitis C virus [14];
Bacterial infections
Helicobacter pylori (HP) untreated and persistent infection has an increased risk of developing a primary gastrointestinal lymphoma [15];
Chlamydia psittaci is responsible for ocular adnexal MALT lymphomas [16];
Borrelia burgdorferi was reported in marginal lymphomas of the skin [17];
Campylobacter jejuni in intestinal lymphomas [18];
Congenital immunodeficiency conditions
Ataxia-telangiectasia;
Wiskott-Aldrich syndrome;
Severe combined immunodeficiency disease;
Acquired immunodeficiency conditions
Chronic immunosuppressive treatment (cytostatic drugs, radiotherapy);
Patients with Human Immunodeficiency Virus (HIV) infection and acquired immunodeficiency syndrome (AIDS) can develop primary central nervous system lymphomas [19];
Autoimmune diseases
Rheumatoid arthritis;
Sjogren’s syndrome;
Systemic lupus erythematosus;
Hashimoto’s thyroiditis is associated with primary thyroid lymphomas [20];
Occupational factors: benzene, dyes, herbicides, pesticides [21,22,23,24];
Family history of malignant hematological diseases and nutritional factors [25].

Share and Cite

MDPI and ACS Style

Corbeanu, R.I.; Găman, A.M. The Involvement of Oxidative Stress in Non-Hodgkin’s Lymphomas; A Review of the Literature. J. Mind Med. Sci. 2022, 9, 1-15. https://doi.org/10.22543/7674.91.P115

AMA Style

Corbeanu RI, Găman AM. The Involvement of Oxidative Stress in Non-Hodgkin’s Lymphomas; A Review of the Literature. Journal of Mind and Medical Sciences. 2022; 9(1):1-15. https://doi.org/10.22543/7674.91.P115

Chicago/Turabian Style

Corbeanu, Ramona Ingrid, and Amelia Maria Găman. 2022. "The Involvement of Oxidative Stress in Non-Hodgkin’s Lymphomas; A Review of the Literature" Journal of Mind and Medical Sciences 9, no. 1: 1-15. https://doi.org/10.22543/7674.91.P115

APA Style

Corbeanu, R. I., & Găman, A. M. (2022). The Involvement of Oxidative Stress in Non-Hodgkin’s Lymphomas; A Review of the Literature. Journal of Mind and Medical Sciences, 9(1), 1-15. https://doi.org/10.22543/7674.91.P115

Article Metrics

Back to TopTop