Next Article in Journal
Thrombotic Events after COVID-19 Vaccination in the Over-50s: Results from a Population-Based Study in Italy
Next Article in Special Issue
EGFR-Based Targeted Therapy for Colorectal Cancer—Promises and Challenges
Previous Article in Journal
Explaining COVID-19 Vaccine Rejection Using Social Cognitive Theory in Qassim, Saudi Arabia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Probiotics in Counteracting the Role of Neutrophils in Cancer Metastasis

by
Upasana Mangrolia
and
Jabez W. Osborne
*
School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India
*
Author to whom correspondence should be addressed.
Vaccines 2021, 9(11), 1306; https://doi.org/10.3390/vaccines9111306
Submission received: 24 September 2021 / Revised: 26 October 2021 / Accepted: 1 November 2021 / Published: 10 November 2021
(This article belongs to the Special Issue Cancer Immunology Focus: Cellular & Molecular Immunology)

Abstract

:
Neutrophils are known for their role geared towards pathogen clearance by different mechanisms that they initiate, primarily by the release of neutrophil extracellular traps (NETs). However, their immune-surveillance capacity accompanied with plasticity in existing as interchangeable subsets, discovered recently, has revealed their property to contribute to complex cancer pathologies including tumor initiation, growth, angiogenesis and metastasis. Although there is a growing body of evidence suggesting a critical balance between the protumoral and antitumoral neutrophil phenotypes, an in-depth signaling pathway analysis would aid in determination of anticipatory, diagnostic and therapeutic targets. This review presents a comprehensive overview of the potential pathways involved in neutrophil-triggered cancer metastasis and introduces the influence of the microbial load and avenues for probiotic intervention.

1. Introduction

Although malignant tumors are predominated by the presence of cancer cells, they also include a range of normal cells capable of influencing the growth and displacement of cancer cells to distant parts of the body. Interestingly, among these noncancerous cells, immune cells cater to promoting the survival and spread of tumors despite being otherwise educated to eradicate them. The possible underlying reasons include their uncontrolled infiltration in the tumor microenvironment and/or their derailed functioning [1].
The role of the immune system in cancer progression has been receiving extensive attention, especially in the context of neutrophils. Neutrophils are the most abundant white blood cells, and their accumulation in tumor sites has been associated with poor patient prognosis. With the discovery of neutrophil subtypes, their phenotype and role in tumors appear to be partially directed by cancer cells [1].
Thus, comprehensively exploring the mechanism is crucial for understanding the pathophysiological aspects and identifying the subsequent targets. These emerging complex targets call for counter convoluted remedies, one of which can be probiotics. At present, the International Scientific Association for Probiotics and Prebiotics (ISAPP) defines probiotics as “live microorganisms that, when administered in adequate amounts, confer a health benefit on the host”. Neutrophils play a role in cancer metastasis via different pathways (e.g., inflammation, immune cell survival status and modulation, angiogenesis, vascular permeability), which have been interestingly explored independently as probiotic targets. In a comprehensive look in the context of connecting probiotics and neutrophil activity, although limited literature is available at present, broad data on the use of probiotics in intervening in neutrophil-mediated metastasis are presented in this review.

2. Immune System Keeping Homeostasis “In Check”

The neutrophils are a part of the innate immune system which also includes eosinophils, basophils, mast cells, dendritic cells (DCs), natural killer cells (NKs) and macrophages. These function in harmony to mount an initial defense by recognizing and attempting to destroy pathogens and abnormal cells such as cancer cells, while also triggering a danger signal to other cells and tissues [2,3]. Thus, during an upsurge inflammatory response. It is important to acknowledge the bystander effect of cytotoxic chemicals on nonpathogenic tissue, extracellular matrix and immune cells. However, the immune cells, while being attached to infection or tissue injury sites by pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs), can respond to signals from abnormal host cells upon prolonged exposure or delayed restoration. A long-term failure in resolving inflammation after infection can thus result in aberrant immune signaling, gene and protein modifications, extensive tissue damage and ultimately carcinogenesis [4]. Further, the immune system’s capacity to impart “immunoediting” in the course of cancer was presented by Dun et al. in 2004 [5]. According to this theory, the immune response to the neoplasm is formed (asymptomatic) where the immune system eliminates the possible tumorigenic cells [6,7]. This identification and extermination of the tumor cells are termed immune surveillance [8]. However, a prolonged fight may lead to sustained cancer in equilibrium with immune cells, with an emerging favorable environment and absence of any distinct symptoms [7,9,10]. This is followed by the last phase where the immune cells receive instructions from the cancer cells to effectively deregulate, block and escape the immune system and their presence is reflected in progressing damage and clinical detectability. There is an increasing body of evidence supporting the role of immune cells beyond the host’s mechanism of fighting off cancer [9,10,11,12], suggesting the intervention of reprogrammed immune cells in enabling tumor growth [13]. Unlike the other immune cells, such as T lymphocytes, dendritic cells, natural killer cells and macrophages, neutrophils have been only recently considered for their role in cancer and are emerging as an important factor in every stage of cancer.

3. Neutrophils: Short-Lived Intense Players of Immunity

Neutrophils constitute about 80% of the white blood cells but are short-lived, which has masked their role in cancer until recently. Neutrophils emerge from the bone marrow in the terminally differentiated state and circulate in the bloodstream with the primarily known purpose of antimicrobial activity. Neutrophils are phagocytes containing chemicals and proteins in their granules (azurophilic, primary; specific, secondary; gelatinase, tertiary), facilitating the internalization of microbes in phagosomes [14]. Their migration is dependent on a variety of chemoattractants originating from injured tissue, pathogenic stimuli, other immune cells or tumors. These chemoattractants can be broadly classified into four groups, namely chemokines, N-formylated peptides, complement anaphylatoxins and lipids, which interact through G-coupled protein receptors (GPCRs) [15]. Upon arriving at the target site, neutrophils secrete β2-integrin-extruding vesicles near the cell surface [16], enabling their tethering. They further secrete matrix metalloproteinases (MMPs) and other oxidants which degrade the endothelial wall, hence allowing the passage into the inflammatory site [17]. The neutrophil-derived oxidants play a key role in the elimination of infectious agents, but under chronic inflammatory conditions, they can cause damage to the host tissue by damaging the DNA and/or impart epigenetic modifications that result in deregulated gene expression which can extend up to abnormal cell division. At the inflamed tissue site, the neutrophils are activated to secrete the cytokines, defensins and stimulatory factors via degranulation, thereby modulating the local tissue environment and recruiting subsequent immune cell cascade (Figure 1) [17].
Under neutropenic condition, as observed in cancer patients upon chemotherapy and radiotherapy [18], when patients are administered the growth factor granulocyte colony-stimulating factor (G-CSF), it can stimulate the de novo neutrophil generation via bone marrow hematopoiesis [19]. Detailed follow-up in patients treated with G-CSF has revealed the onset of pathological conditions accelerating the metastasis [20,21,22,23].

4. Neutrophils in Orchestrating Cancer

The tumor-associated neutrophils (TANs) are suggested to express either of two morphologies: N1 (antitumor) and N2 (protumorigenic). The cytotoxicity of neutrophils on tumor cells is imparted via ROS or antibody-dependent cellular cytotoxicity which stimulates T cells and dendritic cells, further activating other immune components. The N2 phenotype promotes tumor invasion and metastasis; enforcement of angiogenesis; and expression of epidermal growth factor (EGF), transforming growth factor β (TGF-β), platelet-derived growth factor (PDGF) and vascular endothelial growth factor (VEGF) [24,25]. A similar pattern of expression is also observed in inflammatory conditions, and the tumor microenvironment is inclined towards a chronically active inflammatory condition which provides a more conducive state for tumor growth when compared to the acute state [26]. This is derived from the fact that leukocytes at chronic inflammatory sites produce growth factors that promote proliferation (TNF-α, TGF-β, EGF) [27,28,29,30], along with histamine and heparins [31], ECM-modulating proteases; additionally, tumor cells and activated leukocytes produce ROS in abundance, leading to oxidative stress, thereby stimulating the modulation of genetic and epigenetic factors (Figure 2) [32].
It has been reported that TGF-β induces N2 phenotype in TANs [33,34]; when present near tumors, these TANs are observed to release cytokines capable of initiating a positive feedback loop, thereby recruiting a greater number of neutrophils to the tumor site [35]. These TANs not only promote tumor growth or support invasion by angiogenesis, but also mobilize and accumulate in future tumor metastasis sites, i.e., the “premetastatis niche”, and prepare the microenvironment [36,37], as is explained in detail in further sections.
Additionally, myeloid-derived suppressor cells (MDSCs), a group of pathologically activated and systematically expanded immature myeloid cells, are highlighted as being present in tumor-bearing hosts. The neutrophils constitute a part of the polymorphonuclear MDSC (PMN-MDSC) subset and are known to promote immunosurveillance evasion, premetastatic niche formation, EMT and angiogenesis [38,39,40]. MDSC-type neutrophils can highly express inducible nitric oxide synthase (iNOS) and arginase which are involved in the main mechanism for T cell suppression [41,42]; they also induce T cell apoptosis and block the T cell activation as well as effector functions [41,42]. In breast cancer model and ex vivo studies, NK cells are reported to govern the tumor modulatory effects of the neutrophils, where neutrophils, being tumoricidal, are shown to suppress the tumoricidal activity on NK cells. Here, high neutrophil expression suppressed NK cells and subsequently increased metastasis, indicating that although neutrophils and NK cells both have tumoricidal activity, NK cells have higher tumoricidal activity which can eventually downregulate metastasis. Interestingly, in the absence of NK cells, low and not high neutrophil infiltration promoted metastasis [43]. Further, neutrophil extracellular traps (NETs) have been recently found to play a role in providing a physical barrier that shields the tumor cells from interacting with the nearby antitumor immune cells such as CD8+ T cells and NK cells [44,45].

4.1. Cancer Also Promotes Neutrophil Activation

Signals in the form of granulocyte colony-stimulating factor (G-CSF), produced by several tumors themselves, also play a role in increasing neutrophils and inducing their activation [46,47]. The cancer-associated fibroblasts (CAFs), known for tumorigenic activity, produce a protagonist peptide molecule, amyloid β, which can drive NET release via an ROS-dependent pathway [48]. The hyaluronan produced by tumor cells can interact with TLR-4 receptors on neutrophils and induce long-lived neutrophils, which further promotes contact-dependent cancer cell motility; however, this can be blocked by inhibiting the activation of PI3K/Akt signaling in neutrophils, since PI3K/Akt signaling has been implicated in regulating the proinflammatory cytokine expression, thus presenting a potential therapeutic target [49].

4.2. NETs in Mediating Cancer

Although neutrophils are reported to play a negative role in the aggravated inflammatory process, this has been lately attributed to NETs [50,51,52,53,54]. Neutrophils can release histone-bound nuclear DNA along with cytotoxic granules as NETs. NETs have been identified as an element of the nonspecific immunity that affirms their response by causing the microorganisms to stagnate, thereby preventing their spread and ensuring a high localized concentration of antipathogenic factors. These NETs were first identified by Berger et al., who demonstrated the capacity of tumors to form NETs in Ewing’s sarcoma patients [34]. Neutrophil stimulation with phorbol myristate acetate (PMA) showed a web-like release of DNA structures coated with histones, elastase, cathepsin G and myeloperoxidase (MPO) (Figure 3) [34]. Overall, 20 different proteins are thought to be present in a given NETome. Subsequently, it was shown that NET release was associated with neutrophil rupturing, leading NETosis to be characterized as programmed neutrophil death [51]. Unlike apoptosis and necrosis, NETosis is ROS-dependent, which is a result of reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase [51,55]. However, it has been demonstrated that naturally induced NET formation can occur independent of cell membrane lysis [56,57] and occurs via TLR4 activation [58]. Here, the mitochondrial DNA is released [59]; mitochondrial DNA also lacks, by definition, histones. Detailed studies highlight the positive and negative effects of NETs on tumor cells. The NET component MPO has been reported to inhibit and kill melanoma cancer cells. Histones, an important part of the NETs, are capable of destroying the epithelial cells of the blood vessels that feed the tumors. Further, defensins in the NETs mediate tumor cell lysis, recruit dendritic cells and have antiangiogenic properties [29,30,36,48]. MMP-9 has been previously presented as a modulator of innate immune response in antitumor activity [60] in which MMP-9-dependent neutrophil infiltration surfaced as an antitumor process, which can possibly be via N1 phenotype recruitment. Conversely, the TANs and NETs were exclusively present in patients with metastasis and in patients with relapse after intense chemotherapy. Moreover, NETs promote metastasis by degrading the extracellular matrix (ECM) and releasing protease components such as matrix metalloproteinase 9 (MMP-9) that block tumor apoptosis. MMP-9 potentially has been reported to promote angiogenesis and neovascularization within the tumor. Thus, NETs can overall promote migration, invasion and metastasis, as evidently reported in lung cancer cells [31,32]. Since NETs form a physical barrier between the tumor and immune cells, their potential in inhibiting immune cell defense too must not be overlooked.

4.3. NETs in Metastasis

Since neutrophils are a part of the connective tissue (blood), their migration from one organ to another is not unexpected. Moreover, manipulation of tumors during surgery can result in an increased number of free cancer cells in the blood. Thus, the adhesion of cancer cells to neutrophils is of concern since they can act as carriers, which is in accordance with the previously described ability of the neutrophils to migrate to a distant metastatic niche. Interestingly, neutrophils are observed to exert effects on the most rate-limiting stage of the metastatic process: the organ colonization step [21,61]. A study on mice reported the inhibition of lung cancer cell adhesion to PMA-stimulated neutrophils after treatment with DNases [62]; similar reports have been made for lung and pancreatic cancer [63]. This indicates that the neutrophils and/or NETs present in these distant metastatic sites (microvessels) [64] may capture the circulating cancer cells with DNA strands (an important component of NETs). Similarly, understanding the emerging role of NETs in promoting cancer metastasis can open avenues for a more specific or targeted therapeutic. The potential target-pathways include the role of neutrophils and NETs in epithelial–mesenchymal transition (EMT), cancer cell adhesion and premetastatic niche modulation (inflammation, tumor immune evasion, angiogenesis and vascular permeability and awakening the dormant cancer cells) (Figure 4) [65].

4.3.1. NETs Driving the Epithelial–Mesenchymal Transition

Several reports have ascertained that neoplastic cells inherit the ability to invade the local microenvironment and seed in secondary sites. In their journey, however, they encounter several obstacles, including immune surveillance, nutrient/metabolic stress and crossing the endothelial barrier before seeding at the target site. This requires a facilitated translocation which is armed by the immune cells, among which NETs are an emerging player in metastasis. The NETs isolated from PMA-induced human neutrophils in vitro have been reported to cause loss of epithelial junction, promote the development of mesenchymal phenotype and induce aberrant activation of the Wnt/β-catenin signaling pathway in normal epithelial cells [66]. Similar results in neoplastic epithelial cells and in murine models with higher expression of EMT regulators (Snail, Slug and Zeb) along with EMT stimulants (TGF-β and IL-8) have been observed [67,68]. Thus, the ability of NETs to initiate EMT in normal and neoplastic cells suggests their early contribution to metastasis. This is further affirmed in a report where the invasive and migratory characteristics of breast cancer cells were partially impacted upon DNase treatment [67]. Here, in MCF-7 cells, digestion of NETs with DNase did not significantly impact tumor cell migration or MMP9 gene expression, rendering DNA integrity to be dispensable in the effect of NETs over MCF-7 cells [67]. Further, NET-associated elastase or NET-derived HMGB1 inhibition showed abrogation of the EMT stimulation [68]. Collectively the data suggest the role of NET proteins in the EMT initiation; however, since NET-DNA seems to be involved in the EMT process, DNA might contribute to a different metastatic cascade.

4.3.2. Cancer Cell Adhesion

The cancer cells entering the circulating system upon EMT induction are termed circulating tumor cells (CTCs) and have been reported to be present in high amounts in patients [61]. These cells are under constant pressure from the fluid shear forces, oxidative stress and immunity, resulting in few of them entering the micrometastasis state [61]. The NETs accumulated in distant organs are shown to influence the recruitment of CTCs via chemotaxis, as observed in liver metastasis in breast cancer and colon cancer patients where coiled-coil domain-containing protein 25 (CCDC25), a transmembrane protein, acts as an attractant of the NET-associated chemotactic factors [69] and also initiates the β-parvin–RAC1–CDC42 cascade further supporting cytoskeleton rearrangement and directional migration of tumor cells [69]. In the CTC state, the cancer cells must be recognized and sequestered by NETs as a defense appliance, but conversely, NET entrapment advances CTC invasiveness and metastatic potential [70]. Possible mechanisms experimentally deduced are inflammatory signal activation via NET-TLR4-mediated NF-κB [70], STAT3 and MAPK/p38 pathway [71]; here, TLR4 stimulation induces NF-kB-mediated expression of inflammatory cytokines via STAT3 and MAPK/p38 pathway. Additionally, mechanical entrapment in NET-DNA [62] and integrin-mediated adhesion [72] are also some of the possible tethering mechanisms.

4.3.3. NETs in Preparing the Premetastatic Niche

With the immunological and metabolic vulnerability, organotropism of cancer cells requires a hospitable environment at the target organ sites. This premetastatic niche results cumulatively from tumor cells, host stromal cells and bone marrow-derived cells, with a recent focus being neutrophils in metastatic cases of liver [65] and lung [73], partially mediated via NET-remodeled microenvironment; this is described in Figure 4, at the secondary tumor site.

Inflammation and NETs

Inflammation is among the key signs of the premetastatic niche, and the role of NETs in these sites is emerging evidently. The inflammatory milieu provides a seeding environment for the disseminated tumor cells and promotes subsequent survival and proliferation. In colorectal cancer condition, elevated levels of neutrophils at the primary tumor were correlated with the enhanced NETs in the liver, which also induced proinflammatory cytokine (IL-8, IL-6 and TNF-α) production in trapped cancer cells [74]. The hence released TNF-α has been shown to promote EMT [75]. Overall, the neutrophil–NET–inflammation cycle remains a positively regulated cycle promoting cancer metastasis.

Angiogenesis and Vascular Permeability

Since angiogenesis and vascularization are mediums of nutrients, their enhancement becomes a prerequisite for metastatic colonization. Although evidence on the role of NETs in angiogenesis and vascular permeabilization in the context of cancer is limited, their role in tissue repair [76], pulmonary hypertension [77] and corneal neovascularization [78] has been functionally linked to angiogenesis. The possible mechanisms include endothelial cell activation, i.e., TLR-4/NF-κB signaling [77] or inflammation-induced VEGF upregulation [78,79]. Additionally, NETs have been recognized for their role in clearing senescent endothelial cells [80] and disrupting the endothelial barrier, possibly achieved via neutrophil proteases including elastase and MMP [81,82]. Their role in angiogenesis and vascular permeabilization is overall supportive in cancer metastasis.

4.3.4. NETs in Awakening the Dormant Cancer Cells

The plasticity of cancer cells as reflected in their dormant state assists their evasion from the immune system and radiological radar. The disseminated tumor cells, upon reaching the metastatic site, usually undergo dormancy, but over a period of time, they exit the senescence state and resume functionality resulting in metastatic tumors [83]. While resuming their role, the T cells and NK cells mediate their elimination in a programmed manner, thus preventing their relapse [84]. NETs have been unveiled for their role in dormant cell reactivation, mediated by NET-associated proteases [85].
Apart from identifying neutrophils for their role in cancer, it is equally important to address the question of whether neutrophil and NET inhibition is worth the risk when weighed against the chances of sepsis. Additionally, equally promising infection-controlling agents such as antibiotics are readily available; however, their side effects preclude their rational use. Overall, with advancing times, the treatment methods have been enhanced, and so have the possible side effects. Although the options of targeted therapy and immunotherapy are available, conventional chemotherapy is a requirement in case of advanced and recurrent cancer conditions [86]. The chemotherapeutics are commonly known for their side effects such as nausea, diarrhea and vomiting [87,88]. Such side effects are detrimental in terms of nutritional loss and hampered immune system function leading to a delayed treatment cycle, increasing the suffering period and cost [89,90,91].
There is an increase in evidence-based inclination toward the use of probiotics in relieving cancer pathologies, one being gut microbiome restoration (and pathogen elimination) and the other being immune modulation which can promote cancer inhibition. Despite several studies, the explicit role of NETs in cancer still remains elusive. However, with the key components identified in the NETs, selectively targeting the pathology-promoting factors can be a promising approach. With emerging data, probiotics have been identified to counteract the tumor-promoting factors, irrespective of the study being carried out with NETs as the central target.

5. Probiotics as a Potential Therapeutic

The use of bacteria in cancer immunotherapy can be traced back to 1976, when Bacillus Calmette-Guérin (BCG) was first intravesically instilled in bladder cancer patients and was overall successful [92,93]. However, BCG instillation was further found to have side effects including sepsis, organ failure and even disseminated infections [94,95], thereby requiring the discovery of bacteria with similar activity and lesser side effects. A depletion of the microbiota has been correlated with lowered systemic peptidoglycan concentrations, as deduced by less killing of infectious agents (S. pneumoniae and S. aureus) by bone marrow-derived neutrophils. These data thus showed a mechanism for microbiota-mediated systemic immunomodulation, demonstrating that translocated microbial products and their modulation by probiotics can be beneficial for the host in controlling bone marrow myelopoiesis and thereby enhancing systemic innate immune function [96]. Considering the role of neutrophils in almost all steps of cancer metastasis which is exerted in response to tumor-derived incitements [97], the inhibition of their function by probiotics might be an efficient strategy that impedes metastasis. It is important to understand the relationship between the microbiome and the neutrophils to probe probiotic interventions in the desired direction.

5.1. Overcoming Inflammatory Cytokines with Probiotics

Trained immunity is an outcome of a series of nonspecific insults, preparing for protection under secondary points of infection/alterations [98,99]. Despite the probiotic effects on the neutrophil function, the short life span of the neutrophils raises the question of long-term adaptation of neutrophils to similar conditions. On the contrary, this works in the favor of the system, since the effect and activation can thus be regulated. Early investigations have shown that neutrophils can be primed by cytokines such as IL-6, IL-8 and TNF-α, which are regulated by the microbiome [100,101]. Thus, a healthy microbiome from an early stage in life can promote an incessant granulopoiesis by keeping inflammation under control. Additionally, this microbiome can be assisted by probiotics; for instance, a constituent of polysaccharide peptidoglycan complex on Lactobacillus casei strain Shirota (LcS) has been shown to exert beneficial effects in a murine model of inflammatory bowel disease and colitis-associated cancer through inhibition of IL-6/STAT3 signaling (via TLR4 pathway) [97]. Further, with respect to various innate immune cells, the proinflammatory activity of the neutrophils in terms of tissue infiltration, phagocytosis and NET formation elevates with age (from birth to early adult) and is in particular regulated by the TLR4/MyD88 pathway [102]. This has been correlated with the microbiome population, in a study confirming the effect of microbiota composition on neutrophil properties [102].
Similar to cancer cells, infection by pathogens such as enterotoxigenic Escherichia coli (ETEC) K88 has been observed to induce chemokine-mediated neutrophil migration, possibly via IL-8, GRO-a and ENA-78 release, that further leads to CAC pathology [103]. Interestingly, B. animalis or LGG treatment on Caco-2 cells could result in only a low level of neutrophil migration [104], supporting the possible application of these treatments in limiting inflammation-induced neutrophil recruitment.

5.2. Probiotics and Immune Cell Death

Neutrophils that age gradually not only enter senesce but also impart reduced chemotaxis, exhibiting reduced phagocytic activity and enhanced superoxide generation prior to entering the apoptotic phase in the elderly [105]. Their longer presence can impede the average neutrophil function. This can be overcome with strategic probiotic administration. Daily consumption of B. lactis Bi-07 has been shown to improve the phagocytic activity of monocytes and granulocytes in healthy adults (elderly) [106]; this can be considered as the future basis of considering and designing experiments that explore similarly improved phagocytic activity in neutrophils. Lactobacillus helveticus supplementation in vivo increased activity of neutrophil enzymes NADPH oxidase (NOX2) and MPO and moreover maintained the increased levels for a longer duration of 2 months [107]. Park et al. evidently proved the activity of neutrophil NOX2 in causing invasion-promoting NET formation [108]. The study importantly concludes that although probiotic stimulation provokes neutrophil activity, it does not stimulate the innate immune system upon reaching a threshold. This enhancement in the cellular activities to a stable maximum is important since hyperactivation of the immune system can result in unwanted consequences [107]. In contrast to B. lactis Bi-07, it is equally important to scrutinize individual probiotic strains for their activity on phagocytic activity of neutrophils since L. johnsonii La1, also a probiotic, is reported to reduce the phagocytic activity of the neutrophils [109,110]. However, neutrophils incapable of function-reversibility must commit to the death cycle to improve the general output of the immune system.
Neutrophils are abundant in intestinal inflammatory conditions; they are recruited to the site of inflammation as the primary response and can easily cross the epithelium to enter the intestinal lumen. As mentioned earlier, excessive neutrophil infiltration into the mucosa can potentiate severe tissue damage. This overall indicates the need for neutrophil apoptosis and efferocytosis for mucosal homeostasis [111]. The probiotics have access to directly interact with the immunocompetent cells at the intestine, including within Payer’s patches, crypts and lymph nodes [112]. The probiotics at these sites are potentially capable of modulating apoptosis in immune cells. The inflamed mucosa and Lactobacillus casei, when cocultured, showed lowered IL-6 release, which was found to be in correlation with increased apoptotic lymphocyte proportion [113]. Similarly, L. brevis has been shown to induce immune cell apoptosis [114]. Sustrova et al. have demonstrated the effect of B. bifidum, L. rhamnosus and E. faecium on neutrophils to be apoptosis-inducing [115]. On the contrary, Saxami et al. (2017) showed significant inhibition of Caco-2 cell growth and proliferation upon L. pentosus B281 and L. plantarum B282 treatment; however, these strains, in a mouse model, led to rapid recruitment of immune cells, mostly comprising neutrophils among the leukocytes [116]. These results clearly indicate the species-specific activity of probiotics, strengthening the fact that each probiotic has unique functions, thereby requiring detailed studies.

5.3. Probiotics Influencing Neutrophil-Mediated Metastasis

Neutrophil-mediated metastasis is dependent on various tumor microenvironment factors. In this regard, with limited reports correlating the effects of probiotics on neutrophils, it is conceivable to indirectly link probiotic activity on neutrophils via these tumor microenvironment factors. Shinnoh et al. have reported induced TRAIL release from PMNs upon Clostridium butyricum MIYAIRI 588 (CBM588) (probiotic) administration in bladder cancer. The release mechanism of TRAIL from PMNs is not clear; however, MMP-8 is reported to be a key player in TRAIL release [95] and did not enhance TRAIL synthesis. In a previous study, Clostridium butyricum was reported to increase the number of CD8+ T cells and NK cells in lung cancer patients, indicating their promoted proliferation to a cytotoxic end [117]. The number of NK cells increases with age, but their functionality is lowered [118,119,120]. Intravenous injection of L. casei YIT9018 protected the metastatic melanoma-bearing C57BL/6 mice against pulmonary metastasis, and this was augmented with NK cell activity along with auxiliary lymph node cell cytolysis [121]. Although not studied with respect to neutrophils, the previously mentioned recent study shows the influence of active NK cells on neutrophil activity. In another study, Lactobacillus brevis enriched with selenium nanoparticles was found to be capable of reducing liver metastasis in a metastatic mouse (BALB/c) breast cancer model. The study is of relevance to neutrophils since the immune responses are in the form of elevated IFN-γ and IL-17 cytokine levels and enhancement of NK cell activity [122]. Similarly, L. casei Shirota (LcS) has shown augmentation of NK cell cytotoxicity [123].
Through regulation of neutrophils in terms of production, function and apoptosis, the microbiome and thus the probiotics stand a chance to eventually influence the immune system [124]. Vong et al. (2014) deduced the inhibitory activity of Lactobacillus rhamnosus strain GG against NET formation induced by PMA and Staphylococcus aureus. Lactobacillus rhamnosus strain GG also dampened the ROS by exhibiting antioxidant activity, overall protecting against cell cytotoxicity [125]. As mentioned previously, EMT promotes migratory capacity and invasiveness in cancer cells via stromal cell-derived factor 1 (SDF-1) mediated with its receptor CXCR4 through the Wnt/β-catenin pathway. L. acidophilus NCFM has been reported to exert antimetastatic effects by downregulating CXCR4 expression in the spleen, colon and mesenteric lymph nodes of tumor-bearing mice [126]. As chemoprotective modulators, L. casei CRL431 and L. rhamnosus CRL150 showed the ability to increase the number of immature myeloid progenitors in the bone marrow to allow recovery of myeloid cells after cyclophosphamide treatment in mice [96]. Thereby, probiotic supplementation can influence the adaptive feature development attributed to neutrophils along with other immune cells [127]. The effect of probiotics on neutrophil activity in cancer is summarized in Figure 5.

6. Concluding Remarks and Future Directions

The role of neutrophils and the associated NET formation in disease conditions such as cancer is relatively emerging and assertively requires more attention. This review represents an update on the metastatic front of neutrophil activity and puts forward a comprehensive overview of probiotics as a potential therapeutic approach. Although limited, studies indicate the strain specificity of the probiotics towards neutrophil recruitment, indicating an advanced need for thorough examination of the direct and indirect roles of probiotics in cancer via neutrophils.

Author Contributions

Conceptualization, methodology, formal analysis, investigation, resources, data curation, writing—original draft preparation, writing—review and editing, visualization—U.M. and J.W.O.; supervision—J.W.O. 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.

Acknowledgments

The authors are thankful to the Vellore Institute of Technology, India, for providing access to research papers through library facilities.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Seddon, A.R. Neutrophils and Cancer: Regulation of Cell Death and Epigenetic Pathways; University of Otago: Dunedin, New Zealand, 2020. [Google Scholar]
  2. Raulet, D.H. Interplay of natural killer cells and their receptors with the adaptive immune response. Nat. Immunol. 2004, 5, 996–1002. [Google Scholar] [CrossRef] [PubMed]
  3. de Visser, K.E.; Eichten, A.; Coussens, L.M. Paradoxical roles of the immune system during cancer development. Nat. Rev. Cancer 2006, 6, 24–37. [Google Scholar] [CrossRef] [PubMed]
  4. Fullerton, J.N.; Gilroy, D.W. Resolution of inflammation: A new therapeutic frontier. Nat. Rev. Drug Discov. 2016, 15, 551–567. [Google Scholar] [CrossRef] [PubMed]
  5. Dunn, G.P.; Old, L.J.; Schreiber, R.D. The Immunobiology of Cancer Immunosurveillance and Immunoediting. Immunity 2004, 21, 137–148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Gasser, S.; Raulet, D.H. The DNA Damage Response Arouses the Immune System: Figure 1. Cancer Res. 2006, 66, 3959–3962. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Dunn, G.P.; Old, L.J.; Schreiber, R.D. The Three Es of Cancer Immunoediting. Annu. Rev. Immunol. 2004, 22, 329–360. [Google Scholar] [CrossRef] [PubMed]
  8. Zamarron, B.F.; Chen, W. Dual Roles of Immune Cells and Their Factors in Cancer Development and Progression. Int. J. Biol. Sci. 2011, 7, 651–658. [Google Scholar] [CrossRef] [PubMed]
  9. Pagès, F.; Galon, J.; Dieu-Nosjean, M.-C.; Tartour, E.; Sautès-Fridman, C.; Fridman, W.-H. Immune infiltration in human tumors: A prognostic factor that should not be ignored. Oncogene 2010, 29, 1093–1102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Romano, F.; Uggeri, F.; Nespoli, L.; Gianotti, L.; Garancini, M.; Maternini, M.; Nespoli, A.; Uggeri, F. Gastric Cancer Immunotherapy: An Overview. J. Cancer Ther. 2013, 4, 1018–1036. [Google Scholar] [CrossRef] [Green Version]
  11. Ribatti, D. The concept of immune surveillance against tumors: The first theories. Oncotarget 2017, 8, 7175–7180. [Google Scholar] [CrossRef] [Green Version]
  12. Burnet, M. Cancer--A Biological Approach: I. The Processes Of Control. II. The Significance of Somatic Mutation. BMJ 1957, 1, 779–786. [Google Scholar] [CrossRef]
  13. DeNardo, D.G.; Andreu, P.; Coussens, L.M. Interactions between lymphocytes and myeloid cells regulate pro- versus anti-tumor immunity. Cancer Metastasis Rev. 2010, 29, 309–316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Borregaard, N.; Sørensen, O.E.; Theilgaard-Mönch, K. Neutrophil granules: A library of innate immunity proteins. Trends Immunol. 2007, 28, 340–345. [Google Scholar] [CrossRef] [PubMed]
  15. Petri, B.; Sanz, M.-J. Neutrophil chemotaxis. Cell Tissue Res. 2018, 371, 425–436. [Google Scholar] [CrossRef]
  16. Anderson, D.C.; Springer, T.A. Leukocyte Adhesion Deficiency: An Inherited Defect in the Mac-1, LFA-1, and p150,95 Glycoproteins. Annu. Rev. Med. 1987, 38, 175–194. [Google Scholar] [CrossRef] [PubMed]
  17. Häger, M.; Cowland, J.B.; Borregaard, N. Neutrophil granules in health and disease. J. Intern. Med. 2010, 268, 25–34. [Google Scholar] [CrossRef] [PubMed]
  18. Kuderer, N.M.; Dale, D.C.; Crawford, J.; Cosler, L.E.; Lyman, G.H. Mortality, morbidity, and cost associated with febrile neutropenia in adult cancer patients. Cancer 2006, 106, 2258–2266. [Google Scholar] [CrossRef] [PubMed]
  19. Mehta, H.M.; Malandra, M.; Corey, S.J. G-CSF and GM-CSF in Neutropenia. J. Immunol. 2015, 195, 1341–1349. [Google Scholar] [CrossRef] [PubMed]
  20. Kowanetz, M.; Wu, X.; Lee, J.; Tan, M.; Hagenbeek, T.; Qu, X.; Yu, L.; Ross, J.; Korsisaari, N.; Cao, T.; et al. Granulocyte-colony stimulating factor promotes lung metastasis through mobilization of Ly6G+Ly6C+ granulocytes. Proc. Natl. Acad. Sci. USA 2010, 107, 21248–21255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Wculek, S.K.; Malanchi, I. Neutrophils support lung colonization of metastasis-initiating breast cancer cells. Nature 2015, 528, 413–417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. El Rayes, T.; Catena, R.; Lee, S.; Stawowczyk, M.; Joshi, N.; Fischbach, C.; Powell, C.A.; Dannenberg, A.J.; Altorki, N.K.; Gao, D.; et al. Lung inflammation promotes metastasis through neutrophil protease-mediated degradation of Tsp-1. Proc. Natl. Acad. Sci. USA 2015, 112, 16000–16005. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Coffelt, S.B.; Kersten, K.; Doornebal, C.W.; Weiden, J.; Vrijland, K.; Hau, C.-S.; Verstegen, N.J.M.; Ciampricotti, M.; Hawinkels, L.J.A.C.; Jonkers, J.; et al. IL-17-producing γδ T cells and neutrophils conspire to promote breast cancer metastasis. Nature 2015, 522, 345–348. [Google Scholar] [CrossRef] [PubMed]
  24. Granot, Z.; Jablonska, J. Distinct Functions of Neutrophil in Cancer and Its Regulation. Mediat. Inflamm. 2015, 2015, 701067. [Google Scholar] [CrossRef] [Green Version]
  25. Fridlender, Z.G.; Albelda, S.M. Tumor-associated neutrophils: Friend or foe? Carcinogenesis 2012, 33, 949–955. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Balkwill, F.; Charles, K.A.; Mantovani, A. Smoldering and polarized inflammation in the initiation and promotion of malignant disease. Cancer Cell 2005, 7, 211–217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Metzler, K.D.; Fuchs, T.A.; Nauseef, W.M.; Reumaux, D.; Roesler, J.; Schulze, I.; Wahn, V.; Papayannopoulos, V.; Zychlinsky, A. Myeloperoxidase is required for neutrophil extracellular trap formation: Implications for innate immunity. Blood 2011, 117, 953–959. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Al-Benna, S.; Shai, Y.; Jacobsen, F.; Steinstraesser, L. Oncolytic Activities of Host Defense Peptides. Int. J. Mol. Sci. 2011, 12, 8027–8051. [Google Scholar] [CrossRef] [Green Version]
  29. Acuff, H.B.; Carter, K.J.; Fingleton, B.; Gorden, D.L.; Matrisian, L.M. Matrix Metalloproteinase-9 from Bone Marrow–Derived Cells Contributes to Survival but not Growth of Tumor Cells in the Lung Microenvironment. Cancer Res. 2006, 66, 259–266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Masson, V.; De La Ballina, L.R.; Munaut, C.; Wielockx, B.; Jost, M.; Maillard, C.; Blacher, S.; Bajou, K.; Itoh, T.; Itohara, S.; et al. Contribution of host MMP-2 and MMP-9 to promote tumor vascularization and invasion of malignant keratinocytes. FASEB J. 2005, 19, 1–17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Rigoni, A.; Colombo, M.P.; Pucillo, C. The Role of Mast Cells in Molding the Tumor Microenvironment. Cancer Microenviron. 2015, 8, 167–176. [Google Scholar] [CrossRef] [Green Version]
  32. Valko, M.; Leibfritz, D.; Moncol, J.; Cronin, M.T.D.; Mazur, M.; Telser, J. Free radicals and antioxidants in normal physiological functions and human disease. Int. J. Biochem. Cell Biol. 2007, 39, 44–84. [Google Scholar] [CrossRef]
  33. Fridlender, Z.G.; Sun, J.; Kim, S.; Kapoor, V.; Cheng, G.; Ling, L.; Worthen, G.S.; Albelda, S.M. Polarization of Tumor-Associated Neutrophil Phenotype by TGF-β: “N1” versus “N2” TAN. Cancer Cell 2009, 16, 183–194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Berger-Achituv, S.; Brinkmann, V.; Abed, U.A.; Kühn, L.I.; Ben-Ezra, J.; Elhasid, R.; Zychlinsky, A. A proposed role for neutrophil extracellular traps in cancer immunoediting. Front. Immunol. 2013, 4, 48. [Google Scholar] [CrossRef] [Green Version]
  35. Galdiero, M.R.; Bonavita, E.; Barajon, I.; Garlanda, C.; Mantovani, A.; Jaillon, S. Tumor associated macrophages and neutrophils in cancer. Immunobiology 2013, 218, 1402–1410. [Google Scholar] [CrossRef]
  36. Li, L.; Neaves, W.B. Normal Stem Cells and Cancer Stem Cells: The Niche Matters: Figure 1. Cancer Res. 2006, 66, 4553–4557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Wels, J.; Kaplan, R.N.; Rafii, S.; Lyden, D. Migratory neighbors and distant invaders: Tumor-associated niche cells. Genes Dev. 2008, 22, 559–574. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Fleming, V.; Hu, X.; Weber, R.; Nagibin, V.; Groth, C.; Altevogt, P.; Utikal, J.; Umansky, V. Targeting Myeloid-Derived Suppressor Cells to Bypass Tumor-Induced Immunosuppression. Front. Immunol. 2018, 9, 398. [Google Scholar] [CrossRef] [PubMed]
  39. Li, C.; Liu, T.; Bazhin, A.V.; Yang, Y. The Sabotaging Role of Myeloid Cells in Anti-Angiogenic Therapy: Coordination of Angiogenesis and Immune Suppression by Hypoxia. J. Cell. Physiol. 2017, 232, 2312–2322. [Google Scholar] [CrossRef] [PubMed]
  40. Karakhanova, S.; Link, J.; Heinrich, M.; Shevchenko, I.; Yang, Y.; Hassenpflug, M.; Bunge, H.; von Ahn, K.; Brecht, R.; Mathes, A.; et al. Characterization of myeloid leukocytes and soluble mediators in pancreatic cancer: Importance of myeloid-derived suppressor cells. Oncoimmunology 2015, 4, e998519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Youn, J.-I.; Nagaraj, S.; Collazo, M.; Gabrilovich, D.I. Subsets of Myeloid-Derived Suppressor Cells in Tumor-Bearing Mice. J. Immunol. 2008, 181, 5791–5802. [Google Scholar] [CrossRef]
  42. Zea, A.H.; Rodriguez, P.C.; Atkins, M.B.; Hernandez, C.; Signoretti, S.; Zabaleta, J.; McDermott, D.; Quiceno, D.; Youmans, A.; O’Neill, A.; et al. Arginase-Producing Myeloid Suppressor Cells in Renal Cell Carcinoma Patients: A Mechanism of Tumor Evasion. Cancer Res. 2005, 65, 3044–3048. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Li, P.; Lu, M.; Shi, J.; Hua, L.; Gong, Z.; Li, Q.; Shultz, L.D.; Ren, G. Dual roles of neutrophils in metastatic colonization are governed by the host NK cell status. Nat. Commun. 2020, 11, 4387. [Google Scholar] [CrossRef] [PubMed]
  44. Teijeira, Á.; Garasa, S.; Gato, M.; Alfaro, C.; Migueliz, I.; Cirella, A.; de Andrea, C.; Ochoa, M.C.; Otano, I.; Etxeberria, I.; et al. CXCR1 and CXCR2 Chemokine Receptor Agonists Produced by Tumors Induce Neutrophil Extracellular Traps that Interfere with Immune Cytotoxicity. Immunity 2020, 52, 856–871.e8. [Google Scholar] [CrossRef]
  45. Ireland, A.S.; Oliver, T.G. Neutrophils Create an ImpeNETrable Shield between Tumor and Cytotoxic Immune Cells. Immunity 2020, 52, 729–731. [Google Scholar] [CrossRef]
  46. Demers, M.; Krause, D.S.; Schatzberg, D.; Martinod, K.; Voorhees, J.R.; Fuchs, T.A.; Scadden, D.T.; Wagner, D.D. Cancers predispose neutrophils to release extracellular DNA traps that contribute to cancer-associated thrombosis. Proc. Natl. Acad. Sci. USA 2012, 109, 13076–13081. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Demers, M.; Wagner, D.D. Neutrophil extracellular traps. Oncoimmunology 2013, 2, e22946. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Munir, H.; Jones, J.O.; Janowitz, T.; Hoffmann, M.; Euler, M.; Martins, C.P.; Welsh, S.J.; Shields, J.D. Stromal-driven and Amyloid β-dependent induction of neutrophil extracellular traps modulates tumor growth. Nat. Commun. 2021, 12, 683. [Google Scholar] [CrossRef] [PubMed]
  49. Wu, Y.; Zhao, Q.; Peng, C.; Sun, L.; Li, X.-F.; Kuang, D.-M. Neutrophils promote motility of cancer cells via a hyaluronan-mediated TLR4/PI3K activation loop. J. Pathol. 2011, 225, 438–447. [Google Scholar] [CrossRef] [PubMed]
  50. Brinkmann, V. Neutrophil Extracellular Traps Kill Bacteria. Science 2004, 303, 1532–1535. [Google Scholar] [CrossRef] [PubMed]
  51. Fuchs, T.A.; Abed, U.; Goosmann, C.; Hurwitz, R.; Schulze, I.; Wahn, V.; Weinrauch, Y.; Brinkmann, V.; Zychlinsky, A. Novel cell death program leads to neutrophil extracellular traps. J. Cell Biol. 2007, 176, 231–241. [Google Scholar] [CrossRef]
  52. Papayannopoulos, V.; Zychlinsky, A. NETs: A new strategy for using old weapons. Trends Immunol. 2009, 30, 513–521. [Google Scholar] [CrossRef]
  53. von Köckritz-Blickwede, M.; Nizet, V. Innate immunity turned inside-out: Antimicrobial defense by phagocyte extracellular traps. J. Mol. Med. 2009, 87, 775–783. [Google Scholar] [CrossRef] [Green Version]
  54. Abi Abdallah, D.S.; Lin, C.; Ball, C.J.; King, M.R.; Duhamel, G.E.; Denkers, E.Y. Toxoplasma gondii Triggers Release of Human and Mouse Neutrophil Extracellular Traps. Infect. Immun. 2012, 80, 768–777. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Steinberg, B.E.; Grinstein, S. Unconventional Roles of the NADPH Oxidase: Signaling, Ion Homeostasis, and Cell Death. Sci. STKE 2007, 2007, pe11. [Google Scholar] [CrossRef] [PubMed]
  56. Clark, S.R.; Ma, A.C.; Tavener, S.A.; McDonald, B.; Goodarzi, Z.; Kelly, M.M.; Patel, K.D.; Chakrabarti, S.; McAvoy, E.; Sinclair, G.D.; et al. Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood. Nat. Med. 2007, 13, 463–469. [Google Scholar] [CrossRef] [PubMed]
  57. Pilsczek, F.H.; Salina, D.; Poon, K.K.H.; Fahey, C.; Yipp, B.G.; Sibley, C.D.; Robbins, S.M.; Green, F.H.Y.; Surette, M.G.; Sugai, M.; et al. A Novel Mechanism of Rapid Nuclear Neutrophil Extracellular Trap Formation in Response to Staphylococcus aureus. J. Immunol. 2010, 185, 7413–7425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. McDonald, B.; Urrutia, R.; Yipp, B.G.; Jenne, C.N.; Kubes, P. Intravascular Neutrophil Extracellular Traps Capture Bacteria from the Bloodstream during Sepsis. Cell Host Microbe 2012, 12, 324–333. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Yousefi, S.; Mihalache, C.; Kozlowski, E.; Schmid, I.; Simon, H.U. Viable neutrophils release mitochondrial DNA to form neutrophil extracellular traps. Cell Death Differ. 2009, 16, 1438–1444. [Google Scholar] [CrossRef] [PubMed]
  60. Leifler, K.S.; Svensson, S.; Abrahamsson, A.; Bendrik, C.; Robertson, J.; Gauldie, J.; Olsson, A.; Dabrosin, C. Inflammation Induced by MMP-9 Enhances Tumor Regression of Experimental Breast Cancer. J. Immunol. 2013, 190, 4420–4430. [Google Scholar] [CrossRef]
  61. Massagué, J.; Obenauf, A.C. Metastatic colonization by circulating tumour cells. Nature 2016, 529, 298–306. [Google Scholar] [CrossRef]
  62. Cools-Lartigue, J.; Spicer, J.; McDonald, B.; Gowing, S.; Chow, S.; Giannias, B.; Bourdeau, F.; Kubes, P.; Ferri, L. Neutrophil extracellular traps sequester circulating tumor cells and promote metastasis. J. Clin. Investig. 2013, 123, 3446–3458. [Google Scholar] [CrossRef] [PubMed]
  63. Hawes, M.C.; Wen, F.; Elquza, E. Extracellular DNA: A Bridge to Cancer. Cancer Res. 2015, 75, 4260–4264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Garley, M.; Jabłońska, E.; Dąbrowska, D. NETs in cancer. Tumor Biol. 2016, 37, 14355–14361. [Google Scholar] [CrossRef] [PubMed]
  65. Yang, D.; Liu, J. Neutrophil Extracellular Traps: A New Player in Cancer Metastasis and Therapeutic Target. J. Exp. Clin. Cancer Res. 2021, 40, 233. [Google Scholar] [CrossRef] [PubMed]
  66. Pieterse, E.; Rother, N.; Garsen, M.; Hofstra, J.M.; Satchell, S.C.; Hoffmann, M.; Loeven, M.A.; Knaapen, H.K.; van der Heijden, O.W.H.; Berden, J.H.M.; et al. Neutrophil Extracellular Traps Drive Endothelial-to-Mesenchymal Transition. Arterioscler. Thromb. Vasc. Biol. 2017, 37, 1371–1379. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Martins-Cardoso, K.; Almeida, V.H.; Bagri, K.M.; Rossi, M.I.D.; Mermelstein, C.S.; König, S.; Monteiro, R.Q. Neutrophil extracellular traps (Nets) promote pro-metastatic phenotype in human breast cancer cells through epithelial–mesenchymal transition. Cancers 2020, 12, 1542. [Google Scholar] [CrossRef]
  68. Kajioka, H.; Kagawa, S.; Ito, A.; Yoshimoto, M.; Sakamoto, S.; Kikuchi, S.; Kuroda, S.; Yoshida, R.; Umeda, Y.; Noma, K.; et al. Targeting neutrophil extracellular traps with thrombomodulin prevents pancreatic cancer metastasis. Cancer Lett. 2021, 497, 1–13. [Google Scholar] [CrossRef] [PubMed]
  69. Yang, L.; Liu, Q.; Zhang, X.; Liu, X.; Zhou, B.; Chen, J.; Huang, D.; Li, J.; Li, H.; Chen, F.; et al. DNA of neutrophil extracellular traps promotes cancer metastasis via CCDC25. Nature 2020, 583, 133–138. [Google Scholar] [CrossRef]
  70. Yang, L.-Y.; Luo, Q.; Lu, L.; Zhu, W.-W.; Sun, H.-T.; Wei, R.; Lin, Z.-F.; Wang, X.-Y.; Wang, C.-Q.; Lu, M.; et al. Increased neutrophil extracellular traps promote metastasis potential of hepatocellular carcinoma via provoking tumorous inflammatory response. J. Hematol. Oncol. 2020, 13, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Nie, M.; Yang, L.; Bi, X.; Wang, Y.; Sun, P.; Yang, H.; Liu, P.; Li, Z.; Xia, Y.; Jiang, W. Neutrophil Extracellular Traps Induced by IL8 Promote Diffuse Large B-cell Lymphoma Progression via the TLR9 Signaling. Clin. Cancer Res. 2019, 25, 1867–1879. [Google Scholar] [CrossRef] [PubMed]
  72. Monti, M.; De Rosa, V.; Iommelli, F.; Carriero, M.V.; Terlizzi, C.; Camerlingo, R.; Belli, S.; Fonti, R.; Di Minno, G.; Del Vecchio, S. Neutrophil Extracellular Traps as an Adhesion Substrate for Different Tumor Cells Expressing RGD-Binding Integrins. Int. J. Mol. Sci. 2018, 19, 2350. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Rayes, R.F.; Mouhanna, J.G.; Nicolau, I.; Bourdeau, F.; Giannias, B.; Rousseau, S.; Quail, D.; Walsh, L.; Sangwan, V.; Bertos, N.; et al. Primary tumors induce neutrophil extracellular traps with targetable metastasis-promoting effects. JCI Insight 2019, 4, e128008. [Google Scholar] [CrossRef] [Green Version]
  74. Yang, L.; Liu, L.; Zhang, R.; Hong, J.; Wang, Y.; Wang, J.; Zuo, J.; Zhang, J.; Chen, J.; Hao, H. IL-8 mediates a positive loop connecting increased neutrophil extracellular traps (NETs) and colorectal cancer liver metastasis. J. Cancer 2020, 11, 4384–4396. [Google Scholar] [CrossRef] [PubMed]
  75. Bates, R.C.; Mercurio, A.M. Tumor Necrosis Factor-α Stimulates the Epithelial-to-Mesenchymal Transition of Human Colonic Organoids. Mol. Biol. Cell 2003, 14, 1790–1800. [Google Scholar] [CrossRef] [Green Version]
  76. Masuda, H.; Sato, A.; Shizuno, T.; Yokoyama, K.; Suzuki, Y.; Tokunaga, M.; Asahara, T. Batroxobin accelerated tissue repair via neutrophil extracellular trap regulation and defibrinogenation in a murine ischemic hindlimb model. PLoS ONE 2019, 14, e0220898. [Google Scholar] [CrossRef] [Green Version]
  77. Aldabbous, L.; Abdul-Salam, V.; McKinnon, T.; Duluc, L.; Pepke-Zaba, J.; Southwood, M.; Ainscough, A.J.; Hadinnapola, C.; Wilkins, M.R.; Toshner, M.; et al. Neutrophil Extracellular Traps Promote Angiogenesis. Arterioscler. Thromb. Vasc. Biol. 2016, 36, 2078–2087. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Yuan, K.; Zheng, J.; Huang, X.; Zhang, Y.; Han, Y.; Hu, R.; Jin, X. Neutrophil extracellular traps promote corneal neovascularization-induced by alkali burn. Int. Immunopharmacol. 2020, 88, 106902. [Google Scholar] [CrossRef] [PubMed]
  79. Lasch, M.; Kumaraswami, K.; Nasiscionyte, S.; Kircher, S.; van den Heuvel, D.; Meister, S.; Ishikawa-Ankerhold, H.; Deindl, E. RNase A Treatment Interferes With Leukocyte Recruitment, Neutrophil Extracellular Trap Formation, and Angiogenesis in Ischemic Muscle Tissue. Front. Physiol. 2020, 11, 576736. [Google Scholar] [CrossRef] [PubMed]
  80. Binet, F.; Cagnone, G.; Crespo-Garcia, S.; Hata, M.; Neault, M.; Dejda, A.; Wilson, A.M.; Buscarlet, M.; Mawambo, G.T.; Howard, J.P.; et al. Neutrophil extracellular traps target senescent vasculature for tissue remodeling in retinopathy. Science 2020, 369, eaay5356. [Google Scholar] [CrossRef] [PubMed]
  81. Hermant, B.; Bibert, S.; Concord, E.; Dublet, B.; Weidenhaupt, M.; Vernet, T.; Gulino-Debrac, D. Identification of Proteases Involved in the Proteolysis of Vascular Endothelium Cadherin during Neutrophil Transmigration. J. Biol. Chem. 2003, 278, 14002–14012. [Google Scholar] [CrossRef] [PubMed]
  82. Manchanda, K.; Kolarova, H.; Kerkenpaß, C.; Mollenhauer, M.; Vitecek, J.; Rudolph, V.; Kubala, L.; Baldus, S.; Adam, M.; Klinke, A. MPO (Myeloperoxidase) Reduces Endothelial Glycocalyx Thickness Dependent on Its Cationic Charge. Arterioscler. Thromb. Vasc. Biol. 2018, 38, 1859–1867. [Google Scholar] [CrossRef] [PubMed]
  83. Lambert, A.W.; Pattabiraman, D.R.; Weinberg, R.A. Emerging Biological Principles of Metastasis. Cell 2017, 168, 670–691. [Google Scholar] [CrossRef] [Green Version]
  84. Sosa, M.S.; Bragado, P.; Aguirre-Ghiso, J.A. Mechanisms of disseminated cancer cell dormancy: An awakening field. Nat. Rev. Cancer 2014, 14, 611–622. [Google Scholar] [CrossRef] [PubMed]
  85. Albrengues, J.; Shields, M.A.; Ng, D.; Park, C.G.; Ambrico, A.; Poindexter, M.E.; Upadhyay, P.; Uyeminami, D.L.; Pommier, A.; Küttner, V.; et al. Neutrophil extracellular traps produced during inflammation awaken dormant cancer cells in mice. Science 2018, 361, eaao4227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Bradbury, P.; Sivajohanathan, D.; Chan, A.; Kulkarni, S.; Ung, Y.; Ellis, P.M. Postoperative Adjuvant Systemic Therapy in Completely Resected Non-Small-Cell Lung Cancer: A Systematic Review. Clin. Lung Cancer 2017, 18, 259–273.e8. [Google Scholar] [CrossRef] [PubMed]
  87. Andreyev, J.; Ross, P.; Donnellan, C.; Lennan, E.; Leonard, P.; Waters, C.; Wedlake, L.; Bridgewater, J.; Glynne-Jones, R.; Allum, W.; et al. Guidance on the management of diarrhoea during cancer chemotherapy. Lancet. Oncol. 2014, 15, e447-60. [Google Scholar] [CrossRef]
  88. Marx, W.; Kiss, N.; McCarthy, A.L.; McKavanagh, D.; Isenring, L. Chemotherapy-Induced Nausea and Vomiting: A Narrative Review to Inform Dietetics Practice. J. Acad. Nutr. Diet. 2016, 116, 819–827. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Tong, H.; Isenring, E.; Yates, P. The prevalence of nutrition impact symptoms and their relationship to quality of life and clinical outcomes in medical oncology patients. Support. Care Cancer 2009, 17, 83–90. [Google Scholar] [CrossRef] [PubMed]
  90. Tohyama, N.; Tanaka, S.; Onda, K.; Sugiyama, K.; Hirano, T. Influence of anticancer agents on cell survival, proliferation, and CD4+CD25+Foxp3+ regulatory T cell-frequency in human peripheral-blood mononuclear cells activated by T cell-mitogen. Int. Immunopharmacol. 2013, 15, 160–166. [Google Scholar] [CrossRef]
  91. Verma, R.; Foster, R.E.; Horgan, K.; Mounsey, K.; Nixon, H.; Smalle, N.; Hughes, T.A.; Carter, C.R. Lymphocyte depletion and repopulation after chemotherapy for primary breast cancer. Breast Cancer Res. 2016, 18, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Morales, A.; Eidinger, D.; Bruce, A.W. Intracavitary Bacillus Calmette-guerin in the Treatment of Superficial Bladder Tumors. J. Urol. 1976, 116, 180–182. [Google Scholar] [CrossRef]
  93. Herr, H.W.; Morales, A. History of Bacillus Calmette-Guerin and Bladder Cancer: An Immunotherapy Success Story. J. Urol. 2008, 179, 53–56. [Google Scholar] [CrossRef] [PubMed]
  94. Elmer, A.; Bermes, U.; Drath, L.; Büscher, E.; Viertel, A. Sepsis und Multiorganversagen nach BCG-Instillation bei Blasenkarzinom. Internist 2004, 45, 935–939. [Google Scholar] [CrossRef]
  95. Shinnoh, M.; Horinaka, M.; Yasuda, T.; Yoshikawa, S.; Morita, M.; Yamada, T.; Miki, T.; Sakai, T. Clostridium butyricum MIYAIRI 588 shows antitumor effects by enhancing the release of TRAIL from neutrophils through MMP-8. Int. J. Oncol. 2013, 42, 903–911. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Salva, S.; Marranzino, G.; Villena, J.; Agüero, G.; Alvarez, S. Probiotic Lactobacillus strains protect against myelosuppression and immunosuppression in cyclophosphamide-treated mice. Int. Immunopharmacol. 2014, 22, 209–221. [Google Scholar] [CrossRef] [PubMed]
  97. Matsumoto, S.; Hara, T.; Nagaoka, M.; Mike, A.; Mitsuyama, K.; Sako, T.; Yamamoto, M.; Kado, S.; Takada, T. A component of polysaccharide peptidoglycan complex on Lactobacillus induced an improvement of murine model of inflammatory bowel disease and colitis-associated cancer. Immunology 2009, 128, e170–e180. [Google Scholar] [CrossRef] [PubMed]
  98. Netea, M.G.; Joosten, L.A.B.; Latz, E.; Mills, K.H.G.; Natoli, G.; Stunnenberg, H.G.; ONeill, L.A.J.; Xavier, R.J. Trained immunity: A program of innate immune memory in health and disease. Science (80-.) 2016, 352, aaf1098. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Netea, M.G.; Domínguez-Andrés, J.; Barreiro, L.B.; Chavakis, T.; Divangahi, M.; Fuchs, E.; Joosten, L.A.B.; van der Meer, J.W.M.; Mhlanga, M.M.; Mulder, W.J.M.; et al. Defining trained immunity and its role in health and disease. Nat. Rev. Immunol. 2020, 20, 375–388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Ferrante, A. Tumor necrosis factor alpha potentiates neutrophil antimicrobial activity: Increased fungicidal activity against Torulopsis glabrata and Candida albicans and associated increases in oxygen radical production and lysosomal enzyme release. Infect. Immun. 1989, 57, 2115–2122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Yektaei-Karin, E.; Moshfegh, A.; Lundahl, J.; Berggren, V.; Hansson, L.-O.; Marchini, G. The stress of birth enhances in vitro spontaneous and IL-8-induced neutrophil chemotaxis in the human newborn. Pediatr. Allergy Immunol. 2007, 18, 643–651. [Google Scholar] [CrossRef]
  102. Zhang, D.; Chen, G.; Manwani, D.; Mortha, A.; Xu, C.; Faith, J.J.; Burk, R.D.; Kunisaki, Y.; Jang, J.E.; Scheiermann, C.; et al. Neutrophil ageing is regulated by the microbiome. Nature 2015, 525, 528–532. [Google Scholar] [CrossRef] [PubMed]
  103. JAYE, D.L.; PARKOS, C.A. Neutrophil Migration across Intestinal Epithelium. Ann. N. Y. Acad. Sci. 2006, 915, 151–161. [Google Scholar] [CrossRef] [PubMed]
  104. Roselli, M.; Finamore, A.; Britti, M.S.; Mengheri, E. Probiotic bacteria Bifidobacterium animalis MB5 and Lactobacillus rhamnosus GG protect intestinal Caco-2 cells from the inflammation-associated response induced by enterotoxigenic Escherichia coli K88. Br. J. Nutr. 2006, 95, 1177–1184. [Google Scholar] [CrossRef] [PubMed]
  105. Shaw, A.C.; Joshi, S.; Greenwood, H.; Panda, A.; Lord, J.M. Aging of the innate immune system. Curr. Opin. Immunol. 2010, 22, 507–513. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Maneerat, S.; Lehtinen, M.J.; Childs, C.E.; Forssten, S.D.; Alhoniemi, E.; Tiphaine, M.; Yaqoob, P.; Ouwehand, A.C.; Rastall, R.A. Consumption of Bifidobacterium lactis Bi-07 by healthy elderly adults enhances phagocytic activity of monocytes and granulocytes. J. Nutr. Sci. 2013, 2, e44. [Google Scholar] [CrossRef] [Green Version]
  107. apila, R.; Sebastian, R.; Varma, D.V.P.; Sharma, R.; Kapasiya, M.; Salingati, V.; Kapila, S.; Dang, A.K. Comparison of innate immune activation after prolonged feeding of milk fermented with three species of Lactobacilli. Microbiol. Immunol. 2013, 57, 778–784. [Google Scholar] [CrossRef]
  108. Park, J.; Wysocki, R.W.; Amoozgar, Z.; Maiorino, L.; Fein, M.R.; Jorns, J.; Schott, A.F.; Kinugasa-Katayama, Y.; Lee, Y.; Won, N.H.; et al. Cancer cells induce metastasis-supporting neutrophil extracellular DNA traps. Sci. Transl. Med. 2016, 8, 361ra138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Schiffrin, E.J.; Parlesak, A.; Bode, C.; Christian Bode, J.; van’t Hof, M.A.; Grathwohl, D.; Guigoz, Y. Probiotic yogurt in the elderly with intestinal bacterial overgrowth: Endotoxaemia and innate immune functions. Br. J. Nutr. 2008, 101, 961–966. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Miller, L.E.; Lehtoranta, L.; Lehtinen, M.J. The effect of bifidobacterium animalis ssp. lactis HN019 on cellular immune function in healthy elderly subjects: Systematic review and meta-analysis. Nutrients 2017, 9, 191. [Google Scholar] [CrossRef] [PubMed]
  111. Martin, C.J.; Peters, K.N.; Behar, S.M. Macrophages clean up: Efferocytosis and microbial control. Curr. Opin. Microbiol. 2014, 17, 17–23. [Google Scholar] [CrossRef] [Green Version]
  112. Galdeano, C.M.; Perdigón, G. The Probiotic Bacterium Lactobacillus casei Induces Activation of the Gut Mucosal Immune System through Innate Immunity. Clin. Vaccine Immunol. 2006, 13, 219–226. [Google Scholar] [CrossRef] [Green Version]
  113. Carol, M.; Borruel, N.; Antolin, M.; Llopis, M.; Casellas, F.; Guarner, F.; Malagelada, J.-R. Modulation of apoptosis in intestinal lymphocytes by a probiotic bacteria in Crohn’s disease. J. Leukoc. Biol. 2006, 79, 917–922. [Google Scholar] [CrossRef]
  114. Angulo, S.; Morales, A.; Danese, S.; Llacuna, L.; Masamunt, M.C.; Pultz, N.; Cifone, M.G.; De Simone, C.; Delgado, S.; Vila, J.; et al. Probiotic Sonicates Selectively Induce Mucosal Immune Cells Apoptosis through Ceramide Generation via Neutral Sphingomyelinase. PLoS ONE 2011, 6, e16953. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Sustrova, T.; Ondrackova, P.; Leva, L.; Kolarova, M.; Kulich, P.; Sladek, Z. Effect of probiotics on the viability of porcine and human neutrophils in vitro. Vet. Med. (Praha.) 2017, 62, 637–646. [Google Scholar] [CrossRef]
  116. Saxami, G.; Karapetsas, A.; Chondrou, P.; Vasiliadis, S.; Lamprianidou, E.; Kotsianidis, I.; Ypsilantis, P.; Botaitis, S.; Simopoulos, C.; Galanis, A. Potentially probiotic Lactobacillus strains with anti-proliferative activity induce cytokine/chemokine production and neutrophil recruitment in mice. Benef. Microbes 2017, 8, 615–623. [Google Scholar] [CrossRef] [PubMed]
  117. Tian, Y.; Li, M.; Song, W.; Jiang, R.; Li, Y.Q. Effects of probiotics on chemotherapy in patients with lung cancer. Oncol. Lett. 2019, 17, 2836–2848. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Hazeldine, J.; Lord, J.M. The impact of ageing on natural killer cell function and potential consequences for health in older adults. Ageing Res. Rev. 2013, 12, 1069–1078. [Google Scholar] [CrossRef] [PubMed]
  119. Le Garff-Tavernier, M.; Béziat, V.; Decocq, J.; Siguret, V.; Gandjbakhch, F.; Pautas, E.; Debré, P.; Merle-Beral, H.; Vieillard, V. Human NK cells display major phenotypic and functional changes over the life span. Aging Cell 2010, 9, 527–535. [Google Scholar] [CrossRef] [PubMed]
  120. Mocchegiani, E.; Giacconi, R.; Cipriano, C.; Malavolta, M. NK and NKT Cells in Aging and Longevity: Role of Zinc and Metallothioneins. J. Clin. Immunol. 2009, 29, 416–425. [Google Scholar] [CrossRef] [PubMed]
  121. Matsuzaki, T.; Yokokura, T.; Azuma, I. Antimetastatic effect of Lactobacillus casei YIT9018 (LC 9018) on a highly metastatic variant of B16 melanoma in C57BL/6J mice. Cancer Immunol. Immunother. 1987, 24, 99–105. [Google Scholar] [CrossRef] [PubMed]
  122. Wang, W.-J.; Sui, H.; Qi, C.; Li, Q.; Zhang, J.; Wu, S.-F.; Mei, M.-Z.; Lu, Y.-Y.; Wan, Y.-T.; Chang, H.; et al. Ursolic acid inhibits proliferation and reverses drug resistance of ovarian cancer stem cells by downregulating ABCG2 through suppressing the expression of hypoxia-inducible factor-1α in vitro. Oncol. Rep. 2016, 36, 428–440. [Google Scholar] [CrossRef] [PubMed]
  123. Takagi, A.; Matsuzaki, T.; Sato, M.; Nomoto, K.; Morotomi, M.; Yokokura, T. Enhancement of natural killer cytotoxicity delayed murine carcinogenesis by a probiotic microorganism. Carcinogenesis 2001, 22, 599–605. [Google Scholar] [CrossRef]
  124. Lajqi, T.; Pöschl, J.; Frommhold, D.; Hudalla, H. The Role of Microbiota in Neutrophil Regulation and Adaptation in Newborns. Front. Immunol. 2020, 11, 2387. [Google Scholar] [CrossRef]
  125. Vong, L.; Lorentz, R.J.; Assa, A.; Glogauer, M.; Sherman, P.M. Probiotic Lactobacillus rhamnosus Inhibits the Formation of Neutrophil Extracellular Traps. J. Immunol. 2014, 192, 1870–1877. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Keith, B.; Simon, M.C. Hypoxia-Inducible Factors, Stem Cells, and Cancer. Cell 2007, 129, 465–472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Devaux, C.A.; Raoult, D. The Microbiological Memory, an Epigenetic Regulator Governing the Balance Between Good Health and Metabolic Disorders. Front. Microbiol. 2018, 9, 1379. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Neutrophils—from bone marrow to target site: The neutrophils originate from the bone marrow with a short lifespan in circulation. They migrate towards the chemokines following a rolling–tethering–extravasation step. These activated neutrophils both target the infectious agent and further undergo programmed cell death or under tumor-like microenvironment (cancer cells, hypoxia, stroma cell signals) are triggered to undergo NETosis.
Figure 1. Neutrophils—from bone marrow to target site: The neutrophils originate from the bone marrow with a short lifespan in circulation. They migrate towards the chemokines following a rolling–tethering–extravasation step. These activated neutrophils both target the infectious agent and further undergo programmed cell death or under tumor-like microenvironment (cancer cells, hypoxia, stroma cell signals) are triggered to undergo NETosis.
Vaccines 09 01306 g001
Figure 2. Role-dependent differentiation of neutrophils (N1 and N2) in cancer: The polarization of neutrophils is induction-dependent, IFN-β for N1 and TGF-β for N2. N1 neutrophils are categorized as antitumor neutrophils based on their cytotoxicity and T cell activation characteristics. N2 neutrophils are protumor neutrophils that induce carcinogenesis via ROS and RNS pathway; impart cancer growth directed by neutrophil elastases, the MMPs, NETs, NE and IL-1β; and promote cancer cell metastasis. N2 phenotype also suppresses T cell activation and proliferation, induces T cell apoptosis and inhibits NK cell activity; N2-type cells also participate in angiogenesis via CXCR4, MMP9 and VEGF expression modulation.
Figure 2. Role-dependent differentiation of neutrophils (N1 and N2) in cancer: The polarization of neutrophils is induction-dependent, IFN-β for N1 and TGF-β for N2. N1 neutrophils are categorized as antitumor neutrophils based on their cytotoxicity and T cell activation characteristics. N2 neutrophils are protumor neutrophils that induce carcinogenesis via ROS and RNS pathway; impart cancer growth directed by neutrophil elastases, the MMPs, NETs, NE and IL-1β; and promote cancer cell metastasis. N2 phenotype also suppresses T cell activation and proliferation, induces T cell apoptosis and inhibits NK cell activity; N2-type cells also participate in angiogenesis via CXCR4, MMP9 and VEGF expression modulation.
Vaccines 09 01306 g002
Figure 3. Structure of neutrophil extracellular traps: upon receiving the appropriate stimuli, neutrophils release extracellular DNA traps which are overall composed of histone-decorated DNA strands and proteases, including elastase and cathepsin G.
Figure 3. Structure of neutrophil extracellular traps: upon receiving the appropriate stimuli, neutrophils release extracellular DNA traps which are overall composed of histone-decorated DNA strands and proteases, including elastase and cathepsin G.
Vaccines 09 01306 g003
Figure 4. Neutrophil extracellular traps (NETs) induce cancer metastasis: At the primary cancer site, NETs can awaken the dormant cancer cells and can promote epithelial–mesenchymal transition (EMT). Once in the circulation, these circulating cancer cells (CTCs) can become entrapped in the circulating NETs and be protected from immune cells; at the same time, they are exposed to high concentrations of local neutrophil elastase with other factors that can aid in proliferation. The NETs can prepare the secondary/metastatic tumor site, i.e., inhibit immune-surveillance by T cells and NK cells, release MMP-9 and serine proteases which promote angiogenesis via VEGF, modulate extracellular matrix (ECM) in favor of cancer proliferation and recruit inflammatory cytokines which further attract more neutrophils.
Figure 4. Neutrophil extracellular traps (NETs) induce cancer metastasis: At the primary cancer site, NETs can awaken the dormant cancer cells and can promote epithelial–mesenchymal transition (EMT). Once in the circulation, these circulating cancer cells (CTCs) can become entrapped in the circulating NETs and be protected from immune cells; at the same time, they are exposed to high concentrations of local neutrophil elastase with other factors that can aid in proliferation. The NETs can prepare the secondary/metastatic tumor site, i.e., inhibit immune-surveillance by T cells and NK cells, release MMP-9 and serine proteases which promote angiogenesis via VEGF, modulate extracellular matrix (ECM) in favor of cancer proliferation and recruit inflammatory cytokines which further attract more neutrophils.
Vaccines 09 01306 g004
Figure 5. Effect of probiotics on neutrophil activity in cancer condition. Key: red arrows indicate inhibition and green arrows indicate promotion.
Figure 5. Effect of probiotics on neutrophil activity in cancer condition. Key: red arrows indicate inhibition and green arrows indicate promotion.
Vaccines 09 01306 g005
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Mangrolia, U.; Osborne, J.W. Probiotics in Counteracting the Role of Neutrophils in Cancer Metastasis. Vaccines 2021, 9, 1306. https://doi.org/10.3390/vaccines9111306

AMA Style

Mangrolia U, Osborne JW. Probiotics in Counteracting the Role of Neutrophils in Cancer Metastasis. Vaccines. 2021; 9(11):1306. https://doi.org/10.3390/vaccines9111306

Chicago/Turabian Style

Mangrolia, Upasana, and Jabez W. Osborne. 2021. "Probiotics in Counteracting the Role of Neutrophils in Cancer Metastasis" Vaccines 9, no. 11: 1306. https://doi.org/10.3390/vaccines9111306

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