Next Article in Journal
Regenerating Myofibers after an Acute Muscle Injury: What Do We Really Know about Them?
Previous Article in Journal
Management and Mitigation of Vibriosis in Aquaculture: Nanoparticles as Promising Alternatives
Previous Article in Special Issue
Effect of Systemic Inflammation in the CNS: A Silent History of Neuronal Damage
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Crosstalk of Mast Cells and Natural Killer Cells with Neurons in Chemotherapy-Induced Peripheral Neuropathy

1
Hematology, Oncology, Veterans Affairs Long Beach Healthcare System, Long Beach, CA 90822, USA
2
Division of Hematology, Oncology, Department of Medicine, School of Medicine, University of California, Irvine, CA 92617, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(16), 12543; https://doi.org/10.3390/ijms241612543
Submission received: 11 July 2023 / Revised: 4 August 2023 / Accepted: 5 August 2023 / Published: 8 August 2023
(This article belongs to the Special Issue Neuroinflammation Toxicity and Neuroprotection)

Abstract

:
Chemotherapy-induced peripheral neuropathy (CIPN) is a major comorbidity of cancer. Multiple clinical interventions have been studied to effectively treat CIPN, but the results have been disappointing, with no or little efficacy. Hence, understanding the pathophysiology of CIPN is critical to improving the quality of life and clinical outcomes of cancer patients. Although various mechanisms of CIPN have been described in neuropathic anti-cancer agents, the neuroinflammatory process involving cytotoxic/proinflammatory immune cells remains underexamined. While mast cells (MCs) and natural killer (NK) cells are the key innate immune compartments implicated in the pathogenesis of peripheral neuropathy, their role in CIPN has remained under-appreciated. Moreover, the biology of proinflammatory cytokines associated with MCs and NK cells in CIPN is particularly under-evaluated. In this review, we will focus on the interactions between MCs, NK cells, and neuronal structure and their communications via proinflammatory cytokines, including TNFα, IL-1β, and IL-6, in peripheral neuropathy in association with tumor immunology. This review will help lay the foundation to investigate MCs, NK cells, and cytokines to advance future therapeutic strategies for CIPN.

1. Introduction

Chemotherapy-induced peripheral neuropathy (CIPN) is a major comorbidity of cancer. The prevalence of CIPN is estimated at 30–40% of patients treated with neurotoxic agents [1]. Common chemotherapeutic drugs causing CIPN include proteosome inhibitors (e.g., bortezomib) [2], taxanes (e.g., paclitaxel) [3], platinum compounds (e.g., cisplatin) [4], and vinca alkaloids (e.g., vincristine) [5]. In addition, there has been increasing literature data reporting CIPN induced by newer classes of anticancer drugs such as brentuximab vedotin [6] and immune checkpoint inhibitors [7,8]. For example, brentuximab-induced neuropathy was reported in 56% of Hodgkin lymphoma patients, and a meta-analysis of twenty-three clinical trials reported that 4.2% of patients receiving an immune checkpoint inhibitor develop peripheral neuropathy [9]. Unfortunately, CIPN is often the dose-limiting factor of anticancer treatment, which often compromises the chemotherapeutic efficacy, resulting in poor cancer-related clinical outcomes [10,11]. Conversely, the improved survival of cancer patients with advances in cancer therapeutics intensifies the healthcare burden of CIPN [12]. Hence, developing therapeutic strategies for CIPN is critical to improving the quality of life of cancer survivors as well as the outcomes of cancer survival by enhancing tolerability to chemotherapeutics. There have been multiple clinical studies to advance therapeutic interventions including exercise, acupuncture, vitamins, minerals, antidepressants, topical agents, and gabapentinoids for CIPN, but the results have been disappointing with no or limited efficacy [13,14]. For example, gabapentin was not only ineffective in CIPN treatment with little improvement in quality of life [15], but the increased risks of falls and fractures associated with its use have been consistently reported [16,17].
The peripheral sensory network is a highly complex system with multiple non-sensory networks, including immune and non-immune cells such as epidermal keratinocytes, in cross-talk with sensory nerve endings in the skin [18]. An in vitro study using a coculture system of keratinocytes with sensory neurons has demonstrated that keratinocyte membranes directly depolarized by mechanical stimulation can propagate inward currents in the adjacent sensory neurons [19,20]. Keratinocytes are a major inadvertent target of chemotherapeutics, including immune checkpoint inhibitors [21], and cutaneous toxicities are known to be the most common adverse effect of checkpoint inhibitors [22]. Lichenoid dermatitis with dyskeratotic keratinocytes [23] and Steven-Johnson syndrome with apoptotic/necrotic keratinocytes [24] have been reported in patients treated with checkpoint inhibitors blocking CTLA-4, PD-1, and PD-L1, indicating that the unfitness of keratinocytes can be an important mechanism of immunotherapy-induced CIPN.
Although various mechanisms of CIPN have been described in neuropathy-causing anticancer agents, the common pathway involved in CIPN is tightly interdigitated with immunological underpinnings where cytotoxic cells of innate immunity play a key role in propagation of neuroinflammation via neuro-immune synapse formation and release of proinflammatory cytokines [7,13,25]. Although innate immune cells such as mast cells (MCs) and natural killer (NK) cells are profoundly implicated in the pathogenesis of peripheral neuropathy, their roles in CIPN have remained under-appreciated. Moreover, as the current paradigm of oncological therapeutics has shifted from cytotoxic agents to cancer immunotherapy along with targeted therapy [26,27], MCs and NK cells have emerged as the key cellular components in cancer treatment responses and clinical outcomes [1,28,29,30,31]. In this review, we will focus on the interactions between mast cells, NK cells, and neuronal structure in peripheral neuropathy and their association with tumor immunology. This review will help lay the foundation to investigate MCs and NK cells in order to advance future therapeutic strategies for CIPN.
Cytokines are the main humoral vehicle of communication between immune cells, including MCs and NK cells, and neurons. Particularly, proinflammatory cytokines such as TNFα, IL-1β, and IL-6 are well known to play a pivotal role in the progression of CIPN by inducing sensitization of nociceptors and axonal mitochondrial dysfunction and generating reactive oxygen stress in a neuroimmune environment [32]. In this review, we will focus on the interactions between MCs, NK cells, and neural structure and the role of cytokines and soluble factors in peripheral neuropathy in association with tumor immunology. This review will help lay the foundation to investigate MCs, NK cells, and cytokines to advance future therapeutic strategies for CIPN.

2. Mast Cells

Mast cells (MCs) are a part of the innate immune system, acting as the first and fastest responders to pathogens and allergens, and are distributed as tissue-resident myeloid cells primarily in body surfaces exposed to the external environment, including skin tissue and mucosa of the gastrointestinal tract [33,34]. MCs play an important role in both the tumor microenvironment (TME) and the CIPN. MCs are one of the primary innate immune cells in TME, attracted by stem cell factor (SCF) released by tumor cells [35]. MCs orchestrate proinflammatory immune responses by recruiting neutrophils, macrophages, eosinophils, T and B cells, and releasing a variety of inflammatory cytokines, including IL-1, IL-4, IL-6, and TNFα [36]. Moreover, MCs are an important source of angiogenic cytokines such as IL-8, VEGF, and FGF-2 and facilitate tumor invasion and metastasis by producing matrix metalloproteinases (e.g., MMP-2, MMP-9) [28]. Morphine, a narcotic substance commonly used for cancer-related pain, induces tumor progression by increasing tumor angiogenesis and mast cell activation, leading to poorer survival in a transgenic mouse model with breast cancer [37,38]. Recently, MCs in TME were reported to be a detrimental factor for treatment with a checkpoint inhibitor [29]. The presence of intratumoral MCs was associated with a poor immune response to anti-PD-1 therapy, and depletion of MCs by imatinib or sunitinib restored the efficacy of anti-PD-1 therapy, resulting in complete tumor regression in a murine melanoma model [29].
Similar to the detrimental effects of MCs in TME, inflammation induced by MCs plays an important role in peripheral neuropathy [33,39,40]. MCs in the epidermis are colocalized with the nerve terminals of unmyelinated small-diameter C-fibers and myelinated A-delta fibers, key neurons that convey pain stimulation to the trigeminal and dorsal root ganglions and brain [33,41]. The role of MCs in neuropathic pain is well described in a robust murine model of sickle cell anemia, a prototypical disease of severe pain episodes induced by inflammation, oxidative stress, and ischemia-reperfusion injury [42]. Vincent et al. reported that MC inhibition by treatment with cromolyn sodium or imatinib significantly reduces neuroinflammation, and genetic depletion of MCs in sickle cell mice attenuates chronic hypoxia-induced hyperalgesia [43]. Additionally, cannabinoids can alleviate neurogenic inflammation and hyperalgesia while mitigating mast cell activation in sickle cell mice [44], again highlighting the important role of mast cells in painful neuropathy.
MCs excrete or degranulate multiple substances that mediate communication with the neural system. Histamine, an inflammatory substance from MC degranulation, stimulates C fibers to release substance P (SP) [45,46], which is also directly released from MCs and activates adjacent nerve fibers [33]. Conversely, calcitonin gene-related peptide (CGRP), an algogenic substance released from sensory nerves, induces MCs to produce histamine [47]. Not surprisingly, the administration of antihistamine agents is well established to treat and prevent CIPN in clinical practice [48]. Antihistamines along with high-dose dexamethasone are also proven to be efficacious in preventing hypersensitivity (mast cell-mediated acute infusion reactions) induced by oxaliplatin, a platinum agent [49]. Tryptase, a serine proteinase, released from MC granules activates proteinase-activated receptor 2 (PAR-2) [50] that induces neurokinin-1 receptor-dependent hyperalgia [51]. Activation of PAR-2 by tryptase further stimulates afferent neurons to release proinflammatory neuropeptides, including CGRP and SP [52]. (Figure 1A,B) In a murine model, thermal hyperalgesia and tactile allodynia induced by repeated paclitaxel administration correlated with mast cell tryptase activity in peripheral tissue [53]. Treatment with FSLLRY-NH2, a PAR antagonist, or blocking PAR2 downstream signaling, including PLCβ, PKCε, and PKA, diminished paclitaxel-induced neuropathic pain [53]. Another important algogenic mediator released from MCs is sphingosine-1-phosphate (S1P). S1P is the product of sphingosine catalyzed by sphingosine kinases (SphK), activated by crosslinking of FcεRI, IgE receptor [54,55]. S1P binds to its receptors S1P1 and S1P2 in an autocrine manner [55]. S1P mediates mast cell degranulation and migration toward antigens. Furthermore, the pharmacological blockade of S1P on S1P1 mitigated cancer-induced bone pain and neuropathy in a murine model [56], indicating that the S1P pathway can be a major target for CIPN treatment. Additionally, a murine model demonstrated that fingolimod, an S1P1 modulator, attenuates paclitaxel- and oxaliplatin- induced neuropathy and reduces neuroinflammation [57].

3. Natural Killer (NK) Cells

NK cells are cytotoxic lymphocytes of innate immunity. Unlike T cells, lymphocytes of adaptive immunity, NK cells do not require major histocompatibility complex (MHC) restriction for target killing [58]. The interaction between inhibitory killer immunoglobulin-like receptors (KIR) on NK cells and HLA class I molecules on target cells generates inhibitory signals to NK cells via tandem immunoreceptor tyrosine-based inhibitory motifs (ITIMs) [31,59]. NK cells are educated by KIR-HLA interaction, which leads to enhanced NK cell cytotoxicity against foreign, malignant, or virally transformed cells lacking the normal expression of HLA class I (i.e., “missing self”) [60]. The importance of KIR-HLA genotype in the clinical outcomes of cancer treatment has been well described in allogeneic hematopoietic stem cell transplantation (allo-HCT). The KIR genotype can be simply categorized into haplotypes A and B according to the gene contents within the haplotypes. Donors with KIR haplotype B significantly improve disease-free survival in patients with acute myeloid leukemia [61,62,63] and non-Hodgkin lymphoma [64]. Moreover, AML patients’ HLA-C haplotypes confer a significant clinical benefit in allo-HCT [65]. Another potent mechanism of NK cytotoxicity is antibody-dependent cell-mediated cytotoxicity (ADCC). Monoclonal antibodies (moAb) bind to antigens expressed on the surface of tumor cells via the antigen-binding portion (Fab fragment). The other end of moAb is the Fc portion that is recognized FcγRIIIA/CD16a on NK cells. The ligation of CD16a with the Fc portion of the moAb generates potent activating signals to NK cells via the immunoreceptor tyrosine-based activation motif (ITAM) [66,67]. The development of moAb has revolutionized the landscape of cancer treatment [68]. Novel molecules such as bispecific or trispecific killer engagers (i.e., BiKE, TriKE) demonstrated promising antitumor activities by harnessing the ADCC of NK cells [30,69,70,71]. Disintegrin and metalloprotease-17 (ADAM17) expressed on activated NK cells cleaves CD16, which in turn attenuates ADCC activity in NK cells [72]. Inhibition of ADAM17 to prevent CD16 shedding significantly enhanced ADCC mediated by rituximab, a monoclonal anti-CD20 antibody against CD20-expressing tumor cells [72], which indicates that ADCC of NK cells plays a pivotal role in therapeutic efficacy of monoclonal antibodies in cancer treatment. Lastly, the NKG2D pathway is another important mechanism of NK cell activation. Ligands for NKG2D, an activating NK receptor, are often expressed by tumor cells, transformed cells, or infected cells [73]. NKG2D ligands include the RAE (α–ε) encoded by Raet1 genes, H60 (a–c), and MULT1 families in mice and MICA/MICB and ULBPs (1–6) in humans [74,75]. The NKG2D signaling pathway is regarded as the primary host defense mechanism for eradicating “dangerous” cells, as NKG2D ligands can be overexpressed in malignant or infected cells that are subsequently eliminated by NK cells [74]. Shedding MICA/B on tumor cell surfaces and subsequent soluble MICA/B can downregulate NKG2D expression on NK cells, which promotes tumor immune escape by impairing NK cell antitumor activity [76,77]. On the other hand, prolonged activation of NK cells by NKG2D signals results in NK cell exhaustion [78], another potential mechanism of tumor immune evasion. Besides, NK cell cytotoxicity is further determined by the overall balance between inhibitory and activating signals generated by NK cell receptors and ligands expressed on target cells or from soluble factors [31,79,80].
The effect of NK cell cytotoxicity on neuronal degeneration has been described in multiple murine models. In 1982, chronic administration of guanethidine, an adrenergic blocking agent, caused extensive destruction of sympathetic nerves and resulted in “small cell” infiltration [81]. Pretreatment with immunosuppressive agents or irradiation effectively protected against neuronal destruction, indicating that neuronal destruction is mediated by immune mechanisms [81]. Guanethidine administration induced neuronal destruction by infiltration of mononuclear cells even in athymic nude rats, indicating T cell-independent immune-mediated neuronal destruction [82]. A subsequent study revealed that syngeneic IL-2-activated NK cells directly killed embryonal dorsal root ganglia (DRG) neurons via perforin-dependent cytotoxicity, whereas NK cell-mediated lysis was not observed in hippocampal neurons [83]. RAE-1 (an NKG2D ligand) was selectively expressed in embryonal DRG neurons, and anti-NKG2D monoclonal antibodies impaired NK cell-mediated destruction of DRG neurons, which indicates that NKG2D-dependent NK cell cytotoxicity contributes to the degeneration of DRG neurons. NKG2D-dependent NK cell destruction of injured peripheral nerves has been well described [84]. In contrast to embryonal DRG neurons, adult DRG neurons do not express RAE1 as much [84]. However, following axonal injury of peripheral nerves, RAE1 is re-expressed in adult DRG cells, allowing augmented cytotoxicity by activated NK cells via the NKG2D signaling pathway [84]. Although the increased content of granzyme B released from NK cells was identified in injured peripheral nerves, the mere presence of granzyme B in in vitro culture media by separating NK cells from embryonic DRG neurons did not further degenerate the DRG neurons [84]. Hence, the neuro-immune synapse formation of NK cells with nerve fibers is critical for NK cell-induced nerve degeneration. Interestingly, in vivo endogenous NK cytotoxic responses to crushed nerve injury paradoxically reduced chronic neuropathic hypersensitivity in a murine model via the clearance of partially injured sensory axons by activated NK cells [84]. (Figure 1D) Alleviation of neuropathic pain with further degeneration of partially injured exons by NK cytotoxicity is supported by another murine study, where partial sensory fiber loss induced hyperalgesia but more severe axonal loss mitigated hypersensitivity responses [85]. In another murine model of neuropathic pain, electroacupuncture, an effective treatment for neuropathic pain, increased NK cell percentages in the spleen and peripheral blood and NK cell activity measured by the methyl thiazolyl tetrazolium (MTT) assay [86]. However, electroacupuncture treatment did not induce the analgesic effect in mice with in vivo depletion of NK cells, suggesting that NK cells play an important role in the treatment of neuropathic pain [86]. A prospective cohort study reported a significant inverse correlation between the frequency of NK cells in CSF and mechanical pain sensitivity in patients with herpes zoster neuralgia and polyneuropathy (P = 0.004, r = −0.596), indicating a protective role of NK cells in chronic neuropathy [87]. Moreover, the severity of neuropathy inversely correlates with NK cell numbers and NK cell-specific transcription levels in the peripheral blood of patients with chronic inflammatory demyelinating polyneuropathy, again implying the protective role of NK cells in neuropathy [88].

4. Communication between Mast Cells and NK Cells

There is a paucity of data on the interaction between MCs and NK cells specifically in CIPN, although both immune cell groups play significant roles in peripheral neuropathy as described above. However, the communication between MCs and NK cells has been described in other clinical contexts. MCs are shown to attract NK cells via the production of chemokines and cytokines. For example, MCs recruit NK cells to enhance viral clearance during dengue infection [89]. MCs produce CXCL8 in response to reovirus, resulting in NK cell chemotaxis [90] and activating NK cells via the type I interferon response [91]. On the other hand, a hepatocarcinoma murine model demonstrated that tumor-infiltrating mast cells activated by tumor-derived stem cell factor (SCF) augment immunosuppression, and adenosine released by MCs suppresses NK cell activity with the reduction of interferon gamma release in TME [35].
Although NK cells are potent cytotoxic lymphocytes, MCs seem resistant to NK cell cytotoxicity [69,92]. Yun et al. demonstrated that NK cells can successfully irradiate MCs through enhanced ADCC in an in vitro NK cytotoxicity assay [69]. Trispecific killer engager (TriKE), a construct combining a single chain variable fragment (scFv) against CD33 highly expressed on the surface of MCs [93], a scFv against CD16 expressed on NK cells, and IL-15, a key cytokine for NK cell survival, activation, and proliferation, inserted in between as a linker (termed 161533 TriKE), was used to target MCs in this study [69]. 161533 TriKE potently induced NK cell cytotoxicity against MCs, indicating the great therapeutic potential of 161533 TriKE to target MCs by augmenting NK ADCC [69]. (Figure 1C) Investigating the therapeutic potential of 161533 TriKE is warranted in mast cell-associated peripheral neuropathy, especially in the context of CIPN, as NK cell activity is tightly linked to cancer control as well as peripheral neuropathy.

5. Proinflammatory Cytokines

Treatment with cytotoxic chemotherapeutic agents results in cell deaths in neoplastic and normal tissue, which leads to systemic tissue damage and proinflammatory cytokine releases by immune cells (Figure 2) [94,95]. In addition, chemotherapy can directly induce inflammatory responses in the neuronal structure (e.g., dorsal root ganglia) by augmenting the expression of proinflammatory cytokines such as TNFα [96].
As chronic inflammation is a key risk factor for developing various malignancies, inflammatory mediators, including IL-6 and TNFα are known to be involved in carcinogenesis and cancer progression (Figure 2) [97]. Proinflammatory cytokines contribute to generating reactive oxygen species (ROS), and the level of ROS highly correlates with proinflammatory cytokines including TNFα, IL-6, and IL-1β [98,99]. TNFα promotes tumor progression by inducing ROS via cytosolic phospholipase A(2) [100], causing DNA damage, a major carcinogenesis process [101]. Aquaporin (AQP)-3 and AQP-5-mediated diffusion of H2O2, a prototypic ROS molecule, facilitates pancreatic cancer cell migration [102], which characterizes cancer invasion and metastasis. In addition, proinflammatory cytokines serve as growth factors for various types of cancer. For example, metastatic tumor growth can be induced by TNFα release from host hematopoietic cells that mediates NF-κB activation in tumor cells [103]. Activation of the IL-6/STAT3 signaling pathway promotes tumor metastasis [104], and IL-17 released from CD8+ T cells also stimulates cutaneous tumor growth [105]. Multiple in vitro studies have demonstrated that the process of epithelial-to-mesenchymal transition (EMT), a pivotal step for cancer progression with tumor invasion and distant metastasis, is activated by proinflammatory cytokines including TNFα, IL-6, IL-8, and IL-1β [106,107,108,109,110]. ROS also contributes to the EMT process [111], which can be another potential mechanism of proinflammatory cytokine-induced EMT. The protumorigenic effect of inflammation can be mediated by the promotion of angiogenesis, the process of new blood vessel formation to supply oxygen and nutrients to the malignant tissue. Vascular endothelial growth factor (VEGF) is the key molecule for angiogenesis secreted by cancer cells [112]. IL-6-induced STAT3 phosphorylation is associated with increased expression of VEGF and VEGFR2 [113,114]. Moreover, tumor angiogenesis was completely abrogated by a TNFα-neutralizing antibody, indicating that TNFα promotes tumor angiogenesis [115].
MCs are one of the major sources of proinflammatory cytokines. Activated mouse MCs by stem cell factor (SCF) via an interaction with c-kit highly express and release IL-6 [116,117]. MCs produce IL-1 when activated, whereas TNFα is stored in MCs as a preformed mediator [118]. Moreover, MCs can stimulate macrophages to produce IL-1β, as seen in rheumatoid arthritis [119]. Conversely, proinflammatory cytokines affect the function and development of MCs. Based on a murine model, IL-6 and TNFα may contribute to the development of MCs from mast cell precursors [120]. In addition, IL-6 promotes MC survival [121,122]. Under hypoxic conditions, treatment with neutralizing anti-IL-6 antibodies compromised mast cell survival [123]. IL-1, another proinflammatory cytokine, induces IgE-activated MCs to release IL-6 and TNFα along with Th2-related cytokines [124].
Like MCs, NK cell function is tightly associated with proinflammatory cytokines (Figure 2). It is well known that NK cell cytotoxicity is compromised in hyperinflammatory conditions including hemophagocytic lymphohistiocytosis [125], juvenile rheumatoid arthritis, and macrophage activation syndrome [126]. IL-6, primary proinflammatory cytokine implicated in hyperinflammation and cytokine release syndrome [127], impairs NK cell cytotoxicity by downregulating the expression of cytotoxic granules, including perforin and granzyme B [128]. NK cell cytotoxicity can be mediated by TNFα, or TNF-related apoptosis-inducing ligand (TRAIL), a type II membrane protein with homology to TNF [129], which generates apoptotic signaling in the target cells [130,131]. However, TNFα can mediate detrimental effects on NK cell function and survival. Endogenous TNFα induces functional anergy and apoptosis of NK cells activated by triggering CD16 signaling [132]. Proinflammatory cytokines can indirectly compromise NK cell function through ROS induction as well. In an in vitro study, NK cell cytotoxicity was inversely correlated with intracellular ROS production in tumor cells [133]. Moreover, phagocyte-derived ROS impairs NK cell function by diminishing the expression of the NKp46 natural cytotoxicity receptor and NKG2D, an NK cell-activating receptor [134]. ROS-induced NK cell dysfunction can be mediated by A Disintegrin and Metalloprotease 17 (ADAM17), also known as TNFα-converting enzyme (TACE). Oxidative stress and mitochondrial ROS contribute to the increased activity of ADAM17 [135,136]. ADAM17 expressed on NK cells cleaves CD16, leading to the shedding of CD16, the key molecule that mediates ADCC by crosslinking [72,137]. Hence, ROS generated by inflammation can diminish NK cell function by impairing ADCC of NK cells as well as downregulating NK cell-activating receptors.
Neuroinflammation is a common denominator in CIPN, where proinflammatory cytokines serve as messengers for neuro-immune communication [32]. Injury to nervous tissue induces denervated Schwann cells to mount myelomonocytic responses by chemoattraction mediated via leukemia inhibitory factor (LIF) and monocyte chemoattractant protein-1 (MCP-1) in an IL-6-dependent manner [138]. Moreover, activated glial cells following nerve damage produce multiple proinflammatory cytokines, including TNFα, IL-1β, and IL-6 [139,140]. Proinflammatory cytokines not only attract immune cells to the neuroinflammatory tissue but also directly sensitize nociceptors (Figure 2). For example, TNFα and IL-1β can stimulate A- and C- fibers [141]. In rat DRG neurons, the expression of TNF receptors (TNFR1 and TNFR2), TNF-activated p38 mitogen-activated protein kinase (p38MAPK), and c-jun N-terminal kinase (JNC) were observed in immunocytochemical, analysis while TNF-evoked transient increases in [Ca2+] were detected [142]. In a murine model, subcutaneous injection of TNFα resulted in mechanical sensitivity in C nociceptors in a dose-dependent manner, accounting for the generation of hyperalgesia in inflammation [143]. Furthermore, the plantar administration of IL-1β induced a hypersensitive cutaneous reaction to mechanical stimulation [144]. IL-1 signaling impairment significantly reduced pain sensitivity to mechanical and thermal stimulation [145]. Not surprisingly, epidural administration of neutralizing anti-TNFα and anti-IL-1β antibodies markedly diminished pain sensitivity in an additive manner [146]. Although the data had been conflicting, there are multiple clinical studies demonstrating that blockade of proinflammatory cytokines alleviates the symptoms of peripheral neuropathy. A retrospective study demonstrated that perispinal administration of etanercept, a TNFα inhibitor, significantly reduced pain, sensory disturbance, and weakness in patients with treatment-refractory back and neck pain at 1 week, 2 weeks, and 1 month after the treatment [147]. In a randomized, double-blind, placebo-controlled study, the epidural administration of etanercept resulted in significant symptom improvement in patients with sciatica, although this study was limited by a small sample size (n = 24) [148]. In addition to TNFα, targeting IL-6 has been studied in humans. A prospective study comparing the efficacy of epidural administration of tocilizumab, an anti-IL-6 receptor monoclonal antibody, with dexamethasone treatment revealed that tocilizumab treatment was significantly more efficacious for symptom alleviation in patients with lumbar spinal stenosis [149].
Another mechanism by which proinflammatory cytokines trigger neuropathy is the generation of ROS, the main byproduct of proinflammatory cytokines as well as cancer cells [150] and the tumor microenvironment [151]. In fact, chemotherapeutic agents are highly potent in generating ROS, causing multiple tissue damages [152]. The peripheral nervous system is regarded as particularly susceptible to oxidative stress [153]. Naturally, ROS is associated with the development and maintenance of peripheral neuropathy. For example, Mitochondrial ROS production was markedly increased in neuropathic dorsal horns, suggesting that increased neuronal ROS production may be involved in neuropathic sensitization [154], and increased spinal ROS levels due to the production of superoxide from the mitochondria of dorsal horn neurons are associated with maintaining capsaicin-induced hypueralgesia [155,156]. Moreover, inhibition of ROS-by-ROS scavengers including N-tert-Butyl-α-phenylnitrone (PBN) and 4-hydroxy-2,2,6,6-tetramethylpiperidine-1-oxyl (TEMPOL) markedly reduced paclitaxel-induced painful peripheral neuropathy, indicating that ROS plays a central role in the pathogenesis of CIPN [157]. Interestingly, chemotherapy treatment induces an ROS-dependent DNA damage response, which results in upregulation of NK cell ligands on the target cells, leading to NK cell activation [158]. As NK cell-cytotoxicity can exert beneficial therapeutic effects for peripheral neuropathy [84], NK cell-directed treatment can be especially effective in ROS generation-associated CIPN.
It is not surprising that anti-inflammatory cytokines counterbalance proinflammatory cytokines and are associated with beneficial effects in peripheral neuropathy. For example, intrathecal administration of plasmid DNA encoding IL-10 prevented and alleviated paclitaxel-induced peripheral neuropathy in a murine model [159]. Notably, this IL-10-gene therapy resulted in significant reduction in paclitaxel-induced mRNA expression of IL-1β and TNFα in the lumbar DRG, indicating a pivotal role of proinflammatory cytokines in eliciting CIPN. Transforming growth factor β (TGFβ) is another anti-inflammatory cytokine associated with a reduction in neuropathic pain. A murine model demonstrated that intrathecal treatment of TGFβ significantly reduced neuropathic pain by inhibiting the activation of spinal microglia and astrocytes and mitigating spinal inflammatory responses to nerve injury [160]. TGFβ treatment also reduced the expression of IL-1β and IL-6 in the spinal cord with peripheral nerve injury [160].

6. Conclusions

CIPN is a major challenge in cancer treatment. However, the efficacy of therapeutic interventions currently available for CIPN treatment is suboptimal. The biology of CIPN is highly complex. Although MCs and NK cells are known to be highly implicated in the pathogenesis of peripheral neuropathy, there is a paucity of studies on the pathobiology of MCs and NK cells in CIPN. Moreover, the biology of proinflammatory cytokines associated with MCs and NK cells in CIPN is particularly under-evaluated. Based upon the current data, targeting mast cells, proinflammatory cytokines, and/or augmenting the NK cell function in the neuro-immune microenvironment has the potential to improve CIPN. Hence, further studies on the biology of mast cells, NK cells, and their interactions through proinflammatory cytokines in CIPN are warranted.

Author Contributions

Conceptualization, H.D.Y. and K.G.; writing—original draft preparation, H.D.Y.; writing—review and editing, H.D.Y., Y.G. and K.G.; visualization, H.D.Y.; supervision, K.G.; funding acquisition, H.D.Y. All authors have read and agreed to the published version of the manuscript.

Funding

This work is supported by Early-Stage Investigator Award from UCI Chao Family Comprehensive Cancer Center (CFCCC) Anti-Cancer Challenge Pilot Projects 2022 and VA Long Beach Healthcare System Start-up Funding (HDY), and NIH grants CA263806 and HL147562 (KG). The content is solely the responsibility of the authors and does not necessarily represent the official views of the UCI CFCCC, VA Long Beach Healthcare System, and National Institutes of Health.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

Kalpna Gupta received honoraria from Novartis and CSL Behring. Kalpna Gupta’s sources of research grants include Cyclerion, 1910 Genetics, Novartis, Grifols, UCI Foundation, and SCIRE Foundation. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Staff, N.P.; Grisold, A.; Grisold, W.; Windebank, A.J. Chemotherapy-Induced Peripheral Neuropathy: A Current Review. Ann. Neurol. 2017, 81, 772–781. [Google Scholar] [CrossRef]
  2. Richardson, P.G.; Briemberg, H.; Jagannath, S.; Wen, P.Y.; Barlogie, B.; Berenson, J.; Singhal, S.; Siegel, D.S.; Irwin, D.; Schuster, M.; et al. Frequency, Characteristics, and Reversibility of Peripheral Neuropathy during Treatment of Advanced Multiple Myeloma with Bortezomib. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2006, 24, 3113–3120. [Google Scholar] [CrossRef] [PubMed]
  3. Postma, T.J.; Vermorken, J.B.; Liefting, A.J.; Pinedo, H.M.; Heimans, J.J. Paclitaxel-Induced Neuropathy. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 1995, 6, 489–494. [Google Scholar] [CrossRef] [PubMed]
  4. Hammack, J.E.; Michalak, J.C.; Loprinzi, C.L.; Sloan, J.A.; Novotny, P.J.; Soori, G.S.; Tirona, M.T.; Rowland, K.M.; Stella, P.J.; Johnson, J.A. Phase III Evaluation of Nortriptyline for Alleviation of Symptoms of Cis-Platinum-Induced Peripheral Neuropathy. Pain 2002, 98, 195–203. [Google Scholar] [CrossRef]
  5. Kautio, A.-L.; Haanpää, M.; Kautiainen, H.; Kalso, E.; Saarto, T. Burden of Chemotherapy-Induced Neuropathy—A Cross-Sectional Study. Support. Care Cancer Off. J. Multinatl. Assoc. Support. Care Cancer 2011, 19, 1991–1996. [Google Scholar] [CrossRef] [PubMed]
  6. Mariotto, S.; Tecchio, C.; Sorio, M.; Bertolasi, L.; Turatti, M.; Tozzi, M.C.; Benedetti, F.; Cavaletti, G.; Monaco, S.; Ferrari, S. Clinical and Neurophysiological Serial Assessments of Brentuximab Vedotin-Associated Peripheral Neuropathy. Leuk. Lymphoma 2019, 60, 2806–2809. [Google Scholar] [CrossRef] [PubMed]
  7. Oaklander, A.L. Immunotherapy Prospects for Painful Small-Fiber Sensory Neuropathies and Ganglionopathies. Neurother. J. Am. Soc. Exp. Neurother. 2016, 13, 108–117. [Google Scholar] [CrossRef] [Green Version]
  8. Dubey, D.; David, W.S.; Reynolds, K.L.; Chute, D.F.; Clement, N.F.; Cohen, J.V.; Lawrence, D.P.; Mooradian, M.J.; Sullivan, R.J.; Guidon, A.C. Severe Neurological Toxicity of Immune Checkpoint Inhibitors: Growing Spectrum. Ann. Neurol. 2020, 87, 659–669. [Google Scholar] [CrossRef]
  9. Farooq, M.Z.; Aqeel, S.B.; Lingamaneni, P.; Pichardo, R.C.; Jawed, A.; Khalid, S.; Banskota, S.U.; Fu, P.; Mangla, A. Association of Immune Checkpoint Inhibitors With Neurologic Adverse Events: A Systematic Review and Meta-Analysis. JAMA Netw. Open 2022, 5, e227722. [Google Scholar] [CrossRef]
  10. Speck, R.M.; Sammel, M.D.; Farrar, J.T.; Hennessy, S.; Mao, J.J.; Stineman, M.G.; DeMichele, A. Impact of Chemotherapy-Induced Peripheral Neuropathy on Treatment Delivery in Nonmetastatic Breast Cancer. J. Oncol. Pract. 2013, 9, e234–e240. [Google Scholar] [CrossRef]
  11. Gewandter, J.S.; Freeman, R.; Kitt, R.A.; Cavaletti, G.; Gauthier, L.R.; McDermott, M.P.; Mohile, N.A.; Mohlie, S.G.; Smith, A.G.; Tejani, M.A.; et al. Chemotherapy-Induced Peripheral Neuropathy Clinical Trials: Review and Recommendations. Neurology 2017, 89, 859–869. [Google Scholar] [CrossRef] [PubMed]
  12. Kerckhove, N.; Collin, A.; Condé, S.; Chaleteix, C.; Pezet, D.; Balayssac, D. Long-Term Effects, Pathophysiological Mechanisms, and Risk Factors of Chemotherapy-Induced Peripheral Neuropathies: A Comprehensive Literature Review. Front. Pharmacol. 2017, 8, 86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Burgess, J.; Ferdousi, M.; Gosal, D.; Boon, C.; Matsumoto, K.; Marshall, A.; Mak, T.; Marshall, A.; Frank, B.; Malik, R.A.; et al. Chemotherapy-Induced Peripheral Neuropathy: Epidemiology, Pathomechanisms and Treatment. Oncol. Ther. 2021, 9, 385–450. [Google Scholar] [CrossRef]
  14. Loprinzi, C.L.; Lacchetti, C.; Bleeker, J.; Cavaletti, G.; Chauhan, C.; Hertz, D.L.; Kelley, M.R.; Lavino, A.; Lustberg, M.B.; Paice, J.A.; et al. Prevention and Management of Chemotherapy-Induced Peripheral Neuropathy in Survivors of Adult Cancers: ASCO Guideline Update. J. Clin. Oncol. 2020, 38, 3325–3348. [Google Scholar] [CrossRef]
  15. Rao, R.D.; Michalak, J.C.; Sloan, J.A.; Loprinzi, C.L.; Soori, G.S.; Nikcevich, D.A.; Warner, D.O.; Novotny, P.; Kutteh, L.A.; Wong, G.Y.; et al. Efficacy of Gabapentin in the Management of Chemotherapy-Induced Peripheral Neuropathy. Cancer 2007, 110, 2110–2118. [Google Scholar] [CrossRef] [PubMed]
  16. Rentsch, C.T.; Morford, K.L.; Fiellin, D.A.; Bryant, K.J.; Justice, A.C.; Tate, J.P. Safety of Gabapentin Prescribed for Any Indication in a Large Clinical Cohort of 571,718 US Veterans with and without Alcohol Use Disorder. Alcohol Clin. Exp. Res. 2020, 44, 1807–1815. [Google Scholar] [CrossRef]
  17. Ishida, J.H.; McCulloch, C.E.; Steinman, M.A.; Grimes, B.A.; Johansen, K.L. Gabapentin and Pregabalin Use and Association with Adverse Outcomes among Hemodialysis Patients. J. Am. Soc. Nephrol. 2018, 29, 1970. [Google Scholar] [CrossRef] [Green Version]
  18. Moehring, F.; Halder, P.; Seal, R.P.; Stucky, C.L. Uncovering the Cells and Circuits of Touch in Normal and Pathological Settings. Neuron 2018, 100, 349–360. [Google Scholar] [CrossRef] [Green Version]
  19. Moehring, F.; Cowie, A.M.; Menzel, A.D.; Weyer, A.D.; Grzybowski, M.; Arzua, T.; Geurts, A.M.; Palygin, O.; Stucky, C.L. Keratinocytes Mediate Innocuous and Noxious Touch via ATP-P2X4 Signaling. eLife 2018, 7, e31684. [Google Scholar] [CrossRef]
  20. Klusch, A.; Ponce, L.; Gorzelanny, C.; Schäfer, I.; Schneider, S.W.; Ringkamp, M.; Holloschi, A.; Schmelz, M.; Hafner, M.; Petersen, M. Coculture Model of Sensory Neurites and Keratinocytes to Investigate Functional Interaction: Chemical Stimulation and Atomic Force Microscope-Transmitted Mechanical Stimulation Combined with Live-Cell Imaging. J. Investig. Dermatol. 2013, 133, 1387–1390. [Google Scholar] [CrossRef] [Green Version]
  21. Ellis, S.R.; Vierra, A.T.; Millsop, J.W.; Lacouture, M.E.; Kiuru, M. Dermatologic Toxicities to Immune Checkpoint Inhibitor Therapy: A Review of Histopathologic Features. J. Am. Acad. Dermatol. 2020, 83, 1130–1143. [Google Scholar] [CrossRef]
  22. Quach, H.T.; Johnson, D.B.; LeBoeuf, N.R.; Zwerner, J.P.; Dewan, A.K. Cutaneous Adverse Events Caused by Immune Checkpoint Inhibitors. J. Am. Acad. Dermatol. 2021, 85, 956–966. [Google Scholar] [CrossRef] [PubMed]
  23. Sibaud, V. Dermatologic Reactions to Immune Checkpoint Inhibitors: Skin Toxicities and Immunotherapy. Am. J. Clin. Dermatol. 2018, 19, 345–361. [Google Scholar] [CrossRef] [PubMed]
  24. Goldinger, S.M.; Stieger, P.; Meier, B.; Micaletto, S.; Contassot, E.; French, L.E.; Dummer, R. Cytotoxic Cutaneous Adverse Drug Reactions during Anti-PD-1 Therapy. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2016, 22, 4023–4029. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Davies, A.J.; Rinaldi, S.; Costigan, M.; Oh, S.B. Cytotoxic Immunity in Peripheral Nerve Injury and Pain. Front. Neurosci. 2020, 14, 142. [Google Scholar] [CrossRef] [Green Version]
  26. Tsimberidou, A.M.; Fountzilas, E.; Nikanjam, M.; Kurzrock, R. Review of Precision Cancer Medicine: Evolution of the Treatment Paradigm. Cancer Treat. Rev. 2020, 86, 102019. [Google Scholar] [CrossRef]
  27. Pennock, G.K.; Chow, L.Q.M. The Evolving Role of Immune Checkpoint Inhibitors in Cancer Treatment. Oncologist 2015, 20, 812–822. [Google Scholar] [CrossRef] [Green Version]
  28. Ribatti, D.; Crivellato, E. Mast Cells, Angiogenesis, and Tumour Growth. Biochim. Biophys. Acta BBA-Mol. Basis Dis. 2012, 1822, 2–8. [Google Scholar] [CrossRef] [Green Version]
  29. Somasundaram, R.; Connelly, T.; Choi, R.; Choi, H.; Samarkina, A.; Li, L.; Gregorio, E.; Chen, Y.; Thakur, R.; Abdel-Mohsen, M.; et al. Tumor-Infiltrating Mast Cells Are Associated with Resistance to Anti-PD-1 Therapy. Nat. Commun. 2021, 12, 346. [Google Scholar] [CrossRef]
  30. Myers, J.A.; Miller, J.S. Exploring the NK Cell Platform for Cancer Immunotherapy. Nat. Rev. Clin. Oncol. 2021, 18, 85–100. [Google Scholar] [CrossRef]
  31. Foley, B.; Felices, M.; Cichocki, F.; Cooley, S.; Verneris, M.R.; Miller, J.S. The Biology of NK Cells and Their Receptors Affects Clinical Outcomes after Hematopoietic Cell Transplantation (HCT). Immunol. Rev. 2014, 258, 45–63. [Google Scholar] [CrossRef] [PubMed]
  32. Wang, X.-M.; Lehky, T.J.; Brell, J.M.; Dorsey, S.G. Discovering Cytokines as Targets for Chemotherapy-Induced Painful Peripheral Neuropathy. Cytokine 2012, 59, 3–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Gupta, K.; Harvima, I.T. Mast Cell-Neural Interactions Contribute to Pain and Itch. Immunol. Rev. 2018, 282, 168–187. [Google Scholar] [CrossRef] [PubMed]
  34. Krystel-Whittemore, M.; Dileepan, K.N.; Wood, J.G. Mast Cell: A Multi-Functional Master Cell. Front. Immunol. 2015, 6, 620. [Google Scholar] [CrossRef] [Green Version]
  35. Huang, B.; Lei, Z.; Zhang, G.-M.; Li, D.; Song, C.; Li, B.; Liu, Y.; Yuan, Y.; Unkeless, J.; Xiong, H.; et al. SCF-Mediated Mast Cell Infiltration and Activation Exacerbate the Inflammation and Immunosuppression in Tumor Microenvironment. Blood 2008, 112, 1269–1279. [Google Scholar] [CrossRef]
  36. Komi, D.E.A.; Redegeld, F.A. Role of Mast Cells in Shaping the Tumor Microenvironment. Clin. Rev. Allergy Immunol. 2020, 58, 313–325. [Google Scholar] [CrossRef]
  37. Nguyen, J.; Luk, K.; Vang, D.; Soto, W.; Vincent, L.; Robiner, S.; Saavedra, R.; Li, Y.; Gupta, P.; Gupta, K.; et al. Morphine Stimulates Cancer Progression and Mast Cell Activation and Impairs Survival in Transgenic Mice with Breast Cancer. BJA Br. J. Anaesth. 2014, 113, i4–i13. [Google Scholar] [CrossRef] [Green Version]
  38. Novy, D.M.; Nelson, D.V.; Koyyalagunta, D.; Cata, J.P.; Gupta, P.; Gupta, K. Pain, Opioid Therapy, and Survival: A Needed Discussion. Pain 2020, 161, 496–501. [Google Scholar] [CrossRef]
  39. Aich, A.; Afrin, L.B.; Gupta, K. Mast Cell-Mediated Mechanisms of Nociception. Int. J. Mol. Sci. 2015, 16, 29069–29092. [Google Scholar] [CrossRef] [Green Version]
  40. Mittal, A.; Sagi, V.; Gupta, M.; Gupta, K. Mast Cell Neural Interactions in Health and Disease. Front. Cell. Neurosci. 2019, 13, 110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Hendriksen, E.; van Bergeijk, D.; Oosting, R.S.; Redegeld, F.A. Mast Cells in Neuroinflammation and Brain Disorders. Neurosci. Biobehav. Rev. 2017, 79, 119–133. [Google Scholar] [CrossRef] [PubMed]
  42. Tran, H.; Gupta, M.; Gupta, K. Targeting Novel Mechanisms of Pain in Sickle Cell Disease. Blood 2017, 130, 2377–2385. [Google Scholar] [CrossRef] [PubMed]
  43. Vincent, L.; Vang, D.; Nguyen, J.; Gupta, M.; Luk, K.; Ericson, M.E.; Simone, D.A.; Gupta, K. Mast Cell Activation Contributes to Sickle Cell Pathobiology and Pain in Mice. Blood 2013, 122, 1853–1862. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Vincent, L.; Vang, D.; Nguyen, J.; Benson, B.; Lei, J.; Gupta, K. Cannabinoid Receptor-Specific Mechanisms to Alleviate Pain in Sickle Cell Anemia via Inhibition of Mast Cell Activation and Neurogenic Inflammation. Haematologica 2016, 101, 566–577. [Google Scholar] [CrossRef] [Green Version]
  45. Groetzner, P.; Weidner, C. The Human Vasodilator Axon Reflex—An Exclusively Peripheral Phenomenon? PAIN® 2010, 149, 71–75. [Google Scholar] [CrossRef]
  46. Tani, E.; Ishikawa, T. Histamine Acts Directly on Calcitonin Gene-Related Peptide- and Substance P-Containing Trigeminal Ganglion Neurons as Assessed by Calcium Influx and Immunocytochemistry. Auris. Nasus. Larynx 1990, 17, 267–274. [Google Scholar] [CrossRef]
  47. Schwenger, N.; Dux, M.; de Col, R.; Carr, R.; Messlinger, K. Interaction of Calcitonin Gene-Related Peptide, Nitric Oxide and Histamine Release in Neurogenic Blood Flow and Afferent Activation in the Rat Cranial Dura Mater. Cephalalgia Int. J. Headache 2007, 27, 481–491. [Google Scholar] [CrossRef]
  48. Wolf, S.; Barton, D.; Kottschade, L.; Grothey, A.; Loprinzi, C. Chemotherapy-Induced Peripheral Neuropathy: Prevention and Treatment Strategies. Eur. J. Cancer 2008, 44, 1507–1515. [Google Scholar] [CrossRef]
  49. Kidera, Y.; Satoh, T.; Ueda, S.; Okamoto, W.; Okamoto, I.; Fumita, S.; Yonesaka, K.; Hayashi, H.; Makimura, C.; Okamoto, K.; et al. High-Dose Dexamethasone plus Antihistamine Prevents Colorectal Cancer Patients Treated with Modified FOLFOX6 from Hypersensitivity Reactions Induced by Oxaliplatin. Int. J. Clin. Oncol. 2011, 16, 244–249. [Google Scholar] [CrossRef]
  50. Déry, O.; Corvera, C.U.; Steinhoff, M.; Bunnett, N.W. Proteinase-Activated Receptors: Novel Mechanisms of Signaling by Serine Proteases. Am. J. Physiol.-Cell Physiol. 1998, 274, C1429–C1452. [Google Scholar] [CrossRef]
  51. Vergnolle, N.; Bunnett, N.W.; Sharkey, K.A.; Brussee, V.; Compton, S.J.; Grady, E.F.; Cirino, G.; Gerard, N.; Basbaum, A.I.; Andrade-Gordon, P.; et al. Proteinase-Activated Receptor-2 and Hyperalgesia: A Novel Pain Pathway. Nat. Med. 2001, 7, 821–826. [Google Scholar] [CrossRef]
  52. Steinhoff, M.; Vergnolle, N.; Young, S.H.; Tognetto, M.; Amadesi, S.; Ennes, H.S.; Trevisani, M.; Hollenberg, M.D.; Wallace, J.L.; Caughey, G.H.; et al. Agonists of Proteinase-Activated Receptor 2 Induce Inflammation by a Neurogenic Mechanism. Nat. Med. 2000, 6, 151–158. [Google Scholar] [CrossRef] [PubMed]
  53. Chen, Y.; Yang, C.; Wang, Z. Activation of Mast Cell Tryptase and Protease-Activated Receptor 2 Mediate Neuropathic Pain Induced by Paclitaxel. J. Pain 2010, 11, S25. [Google Scholar] [CrossRef]
  54. Prieschl, E.E.; Csonga, R.; Novotny, V.; Kikuchi, G.E.; Baumruker, T. The Balance between Sphingosine and Sphingosine-1-Phosphate Is Decisive for Mast Cell Activation after Fc Epsilon Receptor I Triggering. J. Exp. Med. 1999, 190, 1–8. [Google Scholar] [CrossRef] [PubMed]
  55. Jolly, P.S.; Bektas, M.; Olivera, A.; Gonzalez-Espinosa, C.; Proia, R.L.; Rivera, J.; Milstien, S.; Spiegel, S. Transactivation of Sphingosine-1–Phosphate Receptors by FcεRI Triggering Is Required for Normal Mast Cell Degranulation and Chemotaxis. J. Exp. Med. 2004, 199, 959–970. [Google Scholar] [CrossRef] [PubMed]
  56. Grenald, S.A.; Doyle, T.M.; Zhang, H.; Slosky, L.M.; Chen, Z.; Largent-Milnes, T.M.; Spiegel, S.; Vanderah, T.W.; Salvemini, D. Targeting the S1P/S1PR1 Axis Mitigates Cancer-Induced Bone Pain and Neuroinflammation. Pain 2017, 158, 1733–1742. [Google Scholar] [CrossRef] [Green Version]
  57. Janes, K.; Little, J.W.; Li, C.; Bryant, L.; Chen, C.; Chen, Z.; Kamocki, K.; Doyle, T.; Snider, A.; Esposito, E.; et al. The Development and Maintenance of Paclitaxel-Induced Neuropathic Pain Require Activation of the Sphingosine 1-Phosphate Receptor Subtype 1. J. Biol. Chem. 2014, 289, 21082–21097. [Google Scholar] [CrossRef] [Green Version]
  58. Kärre, K.; Ljunggren, H.G.; Piontek, G.; Kiessling, R. Selective Rejection of H-2-Deficient Lymphoma Variants Suggests Alternative Immune Defence Strategy. Nature 1986, 319, 675–678. [Google Scholar] [CrossRef]
  59. Binstadt, B.A.; Brumbaugh, K.M.; Dick, C.J.; Scharenberg, A.M.; Williams, B.L.; Colonna, M.; Lanier, L.L.; Kinet, J.P.; Abraham, R.T.; Leibson, P.J. Sequential Involvement of Lck and SHP-1 with MHC-Recognizing Receptors on NK Cells Inhibits FcR-Initiated Tyrosine Kinase Activation. Immunity 1996, 5, 629–638. [Google Scholar] [CrossRef] [Green Version]
  60. Guia, S.; Jaeger, B.N.; Piatek, S.; Mailfert, S.; Trombik, T.; Fenis, A.; Chevrier, N.; Walzer, T.; Kerdiles, Y.M.; Marguet, D.; et al. Confinement of Activating Receptors at the Plasma Membrane Controls Natural Killer Cell Tolerance. Sci. Signal. 2011, 4, ra21. [Google Scholar] [CrossRef]
  61. Cooley, S.; Weisdorf, D.J.; Guethlein, L.A.; Klein, J.P.; Wang, T.; Le, C.T.; Marsh, S.G.E.; Geraghty, D.; Spellman, S.; Haagenson, M.D.; et al. Donor Selection for Natural Killer Cell Receptor Genes Leads to Superior Survival after Unrelated Transplantation for Acute Myelogenous Leukemia. Blood 2010, 116, 2411–2419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Cooley, S.; Trachtenberg, E.; Bergemann, T.L.; Saeteurn, K.; Klein, J.; Le, C.T.; Marsh, S.G.E.; Guethlein, L.A.; Parham, P.; Miller, J.S.; et al. Donors with Group B KIR Haplotypes Improve Relapse-Free Survival after Unrelated Hematopoietic Cell Transplantation for Acute Myelogenous Leukemia. Blood 2009, 113, 726–732. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Weisdorf, D.; Cooley, S.; Wang, T.; Trachtenberg, E.; Vierra-Green, C.; Spellman, S.; Sees, J.A.; Spahn, A.; Vogel, J.; Fehniger, T.A.; et al. KIR B Donors Improve the Outcome for AML Patients given Reduced Intensity Conditioning and Unrelated Donor Transplantation. Blood Adv. 2020, 4, 740–754. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Bachanova, V.; Weisdorf, D.J.; Wang, T.; Marsh, S.G.E.; Trachtenberg, E.; Haagenson, M.D.; Spellman, S.R.; Ladner, M.; Guethlein, L.A.; Parham, P.; et al. Donor KIR B Genotype Improves Progression-Free Survival of Non-Hodgkin Lymphoma Patients Receiving Unrelated Donor Transplantation. Biol. Blood Marrow Transplant. 2016, 22, 1602–1607. [Google Scholar] [CrossRef] [Green Version]
  65. Cooley, S.; Weisdorf, D.J.; Guethlein, L.A.; Klein, J.P.; Wang, T.; Marsh, S.G.E.; Spellman, S.; Haagenson, M.D.; Saeturn, K.; Ladner, M.; et al. Donor Killer Cell Ig-like Receptor B Haplotypes, Recipient HLA-C1, and HLA-C Mismatch Enhance the Clinical Benefit of Unrelated Transplantation for Acute Myelogenous Leukemia. J. Immunol. 2014, 192, 4592–4600. [Google Scholar] [CrossRef] [Green Version]
  66. Vivier, E.; Morin, P.; O’Brien, C.; Druker, B.; Schlossman, S.F.; Anderson, P. Tyrosine Phosphorylation of the Fc Gamma RIII(CD16): Zeta Complex in Human Natural Killer Cells. Induction by Antibody-Dependent Cytotoxicity but Not by Natural Killing. J. Immunol. 1991, 146, 206–210. [Google Scholar] [CrossRef]
  67. Nimmerjahn, F.; Ravetch, J.V. Fcγ Receptors as Regulators of Immune Responses. Nat. Rev. Immunol. 2008, 8, 34–47. [Google Scholar] [CrossRef]
  68. Adams, G.P.; Weiner, L.M. Monoclonal Antibody Therapy of Cancer. Nat. Biotechnol. 2005, 23, 1147–1157. [Google Scholar] [CrossRef]
  69. Yun, H.D.; Felices, M.; Vallera, D.A.; Hinderlie, P.; Cooley, S.; Arock, M.; Gotlib, J.; Ustun, C.; Miller, J.S. Trispecific Killer Engager CD16xIL15xCD33 Potently Induces NK Cell Activation and Cytotoxicity against Neoplastic Mast Cells. Blood Adv. 2018, 2, 1580–1584. [Google Scholar] [CrossRef]
  70. Felices, M.; Lenvik, T.R.; Davis, Z.B.; Miller, J.S.; Vallera, D.A. Generation of BiKEs and TriKEs to Improve NK Cell-Mediated Targeting of Tumor Cells. Methods Mol. Biol. Clifton NJ 2016, 1441, 333–346. [Google Scholar] [CrossRef] [Green Version]
  71. Gleason, M.K.; Ross, J.A.; Warlick, E.D.; Lund, T.C.; Verneris, M.R.; Wiernik, A.; Spellman, S.; Haagenson, M.D.; Lenvik, A.J.; Litzow, M.R.; et al. CD16xCD33 Bispecific Killer Cell Engager (BiKE) Activates NK Cells against Primary MDS and MDSC CD33+ Targets. Blood 2014, 123, 3016–3026. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Romee, R.; Foley, B.; Lenvik, T.; Wang, Y.; Zhang, B.; Ankarlo, D.; Luo, X.; Cooley, S.; Verneris, M.; Walcheck, B.; et al. NK Cell CD16 Surface Expression and Function Is Regulated by a Disintegrin and Metalloprotease-17 (ADAM17). Blood 2013, 121, 3599–3608. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Raulet, D.H.; Gasser, S.; Gowen, B.G.; Deng, W.; Jung, H. Regulation of Ligands for the NKG2D Activating Receptor. Annu. Rev. Immunol. 2013, 31, 413–441. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Lanier, L.L. NKG2D Receptor and Its Ligands in Host Defense. Cancer Immunol. Res. 2015, 3, 575–582. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Eagle, R.A.; Trowsdale, J. Promiscuity and the Single Receptor: NKG2D. Nat. Rev. Immunol. 2007, 7, 737–744. [Google Scholar] [CrossRef]
  76. Groh, V.; Wu, J.; Yee, C.; Spies, T. Tumour-Derived Soluble MIC Ligands Impair Expression of NKG2D and T-Cell Activation. Nature 2002, 419, 734–738. [Google Scholar] [CrossRef]
  77. Cichocki, F.; Miller, J.S. Promoting T and NK Cell Attack: Preserving Tumor MICA/B by Vaccines. Cell Res. 2022, 32, 961–962. [Google Scholar] [CrossRef]
  78. Myers, J.A.; Schirm, D.; Bendzick, L.; Hopps, R.; Selleck, C.; Hinderlie, P.; Felices, M.; Miller, J.S. Balanced Engagement of Activating and Inhibitory Receptors Mitigates Human NK Cell Exhaustion. JCI Insight 2022, 7, e150079. [Google Scholar] [CrossRef]
  79. Kennedy, P.R.; Felices, M.; Miller, J.S. Challenges to the Broad Application of Allogeneic Natural Killer Cell Immunotherapy of Cancer. Stem Cell Res. Ther. 2022, 13, 165. [Google Scholar] [CrossRef]
  80. Yun, H.D.; Schirm, D.K.; Felices, M.; Miller, J.S.; Eckfeldt, C.E. Dinaciclib Enhances Natural Killer Cell Cytotoxicity against Acute Myelogenous Leukemia. Blood Adv. 2019, 3, 2448–2452. [Google Scholar] [CrossRef]
  81. Manning, P.T.; Russell, J.H.; Johnson, E.M. Immunosuppressive Agents Prevent Guanethidine-Induced Destruction of Rat Sympathetic Neurons. Brain Res. 1982, 241, 131–143. [Google Scholar] [CrossRef] [PubMed]
  82. Thygesen, P.; Hougen, H.P.; Christensen, H.B.; Rygaard, J.; Svendsen, O.; Juul, P. Identification of the Mononuclear Cell Infiltrate in the Superior Cervical Ganglion of Athymic Nude and Euthymic Rats after Guanethidine-Induced Sympathectomy. Int. J. Immunopharmacol. 1990, 12, 327–330. [Google Scholar] [CrossRef] [PubMed]
  83. Backström, E.; Chambers, B.J.; Kristensson, K.; Ljunggren, H.-G. Direct NK Cell-Mediated Lysis of Syngenic Dorsal Root Ganglia Neurons In Vitro1. J. Immunol. 2000, 165, 4895–4900. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Davies, A.J.; Kim, H.W.; Gonzalez-Cano, R.; Choi, J.; Back, S.K.; Roh, S.E.; Johnson, E.; Gabriac, M.; Kim, M.-S.; Lee, J.; et al. Natural Killer Cells Degenerate Intact Sensory Afferents Following Nerve Injury. Cell 2019, 176, 716–728. [Google Scholar] [CrossRef] [Green Version]
  85. Huang, C.; Zou, W.; Lee, K.; Wang, E.; Zhu, X.; Guo, Q. Different Symptoms of Neuropathic Pain Can Be Induced by Different Degrees of Compressive Force on the C7 Dorsal Root of Rats. Spine J. Off. J. North Am. Spine Soc. 2012, 12, 1154–1160. [Google Scholar] [CrossRef]
  86. Gao, Y.-H.; Wang, J.-Y.; Qiao, L.-N.; Chen, S.-P.; Tan, L.-H.; Xu, Q.-L.; Liu, J.-L. NK Cells Mediate the Cumulative Analgesic Effect of Electroacupuncture in a Rat Model of Neuropathic Pain. BMC Complement. Altern. Med. 2014, 14, 316. [Google Scholar] [CrossRef] [Green Version]
  87. Lassen, J.; Stürner, K.H.; Gierthmühlen, J.; Dargvainiene, J.; Kixmüller, D.; Leypoldt, F.; Baron, R.; Hüllemann, P. Protective Role of Natural Killer Cells in Neuropathic Pain Conditions. PAIN 2021, 162, 2366. [Google Scholar] [CrossRef]
  88. Mausberg, A.K.; Heininger, M.K.; Meyer Zu Horste, G.; Cordes, S.; Fleischer, M.; Szepanowski, F.; Kleinschnitz, C.; Hartung, H.-P.; Kieseier, B.C.; Stettner, M. NK Cell Markers Predict the Efficacy of IV Immunoglobulins in CIDP. Neurol. Neuroimmunol. Neuroinflamm. 2020, 7, e884. [Google Scholar] [CrossRef]
  89. St. John, A.L.; Rathore, A.P.S.; Yap, H.; Ng, M.-L.; Metcalfe, D.D.; Vasudevan, S.G.; Abraham, S.N. Immune Surveillance by Mast Cells during Dengue Infection Promotes Natural Killer (NK) and NKT-Cell Recruitment and Viral Clearance. Proc. Natl. Acad. Sci. USA 2011, 108, 9190–9195. [Google Scholar] [CrossRef]
  90. Burke, S.M.; Issekutz, T.B.; Mohan, K.; Lee, P.W.K.; Shmulevitz, M.; Marshall, J.S. Human Mast Cell Activation with Virus-Associated Stimuli Leads to the Selective Chemotaxis of Natural Killer Cells by a CXCL8-Dependent Mechanism. Blood 2008, 111, 5467–5476. [Google Scholar] [CrossRef] [Green Version]
  91. Portales-Cervantes, L.; Haidl, I.D.; Lee, P.W.; Marshall, J.S. Virus-Infected Human Mast Cells Enhance Natural Killer Cell Functions. J. Innate Immun. 2017, 9, 94–108. [Google Scholar] [CrossRef]
  92. Ustun, C.; Williams, S.; Skendzel, S.; Kodal, B.; Arock, M.; Gotlib, J.; Vallera, D.A.; Cooley, S.; Felices, M.; Weisdorf, D.; et al. Allogeneic NK Cells Eradicate Myeloblasts but Not Neoplastic Mast Cells in Systemic Mastocytosis Associated with Acute Myeloid Leukemia. Am. J. Hematol. 2017, 92, E66–E68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Krauth, M.-T.; Böhm, A.; Agis, H.; Sonneck, K.; Samorapoompichit, P.; Florian, S.; Sotlar, K.; Valent, P. Effects of the CD33-Targeted Drug Gemtuzumab Ozogamicin (Mylotarg) on Growth and Mediator Secretion in Human Mast Cells and Blood Basophils. Exp. Hematol. 2007, 35, 108–116. [Google Scholar] [CrossRef] [PubMed]
  94. Starobova, H.; Vetter, I. Pathophysiology of Chemotherapy-Induced Peripheral Neuropathy. Front. Mol. Neurosci. 2017, 10, 174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Javeed, A.; Ashraf, M.; Riaz, A.; Ghafoor, A.; Afzal, S.; Mukhtar, M.M. Paclitaxel and Immune System. Eur. J. Pharm. Sci. 2009, 38, 283–290. [Google Scholar] [CrossRef] [PubMed]
  96. Oliveira, H.R.; Coelho, M.S.; Neves, F.D.A.R.; Duarte, D.B. Cisplatin-Induced Changes in Calcitonin Gene-Related Peptide or TNF-α Release in Rat Dorsal Root Ganglia in Vitro Model of Neurotoxicity Are Not Reverted by Rosiglitazone. Neurotoxicology 2022, 93, 211–221. [Google Scholar] [CrossRef]
  97. Landskron, G.; De la Fuente, M.; Thuwajit, P.; Thuwajit, C.; Hermoso, M.A. Chronic Inflammation and Cytokines in the Tumor Microenvironment. J. Immunol. Res. 2014, 2014, e149185. [Google Scholar] [CrossRef] [Green Version]
  98. Yang, D.; Elner, S.G.; Bian, Z.-M.; Till, G.O.; Petty, H.R.; Elner, V.M. Pro-Inflammatory Cytokines Increase Reactive Oxygen Species through Mitochondria and NADPH Oxidase in Cultured RPE Cells. Exp. Eye Res. 2007, 85, 462–472. [Google Scholar] [CrossRef] [Green Version]
  99. Mateen, S.; Moin, S.; Shahzad, S.; Khan, A.Q. Level of Inflammatory Cytokines in Rheumatoid Arthritis Patients: Correlation with 25-Hydroxy Vitamin D and Reactive Oxygen Species. PLoS ONE 2017, 12, e0178879. [Google Scholar] [CrossRef]
  100. Woo, C.H.; Eom, Y.W.; Yoo, M.H.; You, H.J.; Han, H.J.; Song, W.K.; Yoo, Y.J.; Chun, J.S.; Kim, J.H. Tumor Necrosis Factor-Alpha Generates Reactive Oxygen Species via a Cytosolic Phospholipase A2-Linked Cascade. J. Biol. Chem. 2000, 275, 32357–32362. [Google Scholar] [CrossRef] [Green Version]
  101. Salehi, F.; Behboudi, H.; Kavoosi, G.; Ardestani, S.K. Oxidative DNA Damage Induced by ROS-Modulating Agents with the Ability to Target DNA: A Comparison of the Biological Characteristics of Citrus Pectin and Apple Pectin. Sci. Rep. 2018, 8, 13902. [Google Scholar] [CrossRef] [Green Version]
  102. Rodrigues, C.; Pimpão, C.; Mósca, A.F.; Coxixo, A.S.; Lopes, D.; da Silva, I.V.; Pedersen, P.A.; Antunes, F.; Soveral, G. Human Aquaporin-5 Facilitates Hydrogen Peroxide Permeation Affecting Adaption to Oxidative Stress and Cancer Cell Migration. Cancers 2019, 11, 932. [Google Scholar] [CrossRef] [Green Version]
  103. Luo, J.-L.; Maeda, S.; Hsu, L.-C.; Yagita, H.; Karin, M. Inhibition of NF-KappaB in Cancer Cells Converts Inflammation- Induced Tumor Growth Mediated by TNFalpha to TRAIL-Mediated Tumor Regression. Cancer Cell 2004, 6, 297–305. [Google Scholar] [CrossRef] [Green Version]
  104. Zheng, T.; Hong, X.; Wang, J.; Pei, T.; Liang, Y.; Yin, D.; Song, R.; Song, X.; Lu, Z.; Qi, S.; et al. Gankyrin Promotes Tumor Growth and Metastasis through Activation of IL-6/STAT3 Signaling in Human Cholangiocarcinoma. Hepatology 2014, 59, 935–946. [Google Scholar] [CrossRef]
  105. He, D.; Li, H.; Yusuf, N.; Elmets, C.A.; Athar, M.; Katiyar, S.K.; Xu, H. IL-17 Mediated Inflammation Promotes Tumor Growth and Progression in the Skin. PLoS ONE 2012, 7, e32126. [Google Scholar] [CrossRef] [Green Version]
  106. Mittal, V. Epithelial Mesenchymal Transition in Tumor Metastasis. Annu. Rev. Pathol. Mech. Dis. 2018, 13, 395–412. [Google Scholar] [CrossRef]
  107. Bates, R.C.; Mercurio, A.M. Tumor Necrosis Factor-Alpha Stimulates the Epithelial-to-Mesenchymal Transition of Human Colonic Organoids. Mol. Biol. Cell 2003, 14, 1790–1800. [Google Scholar] [CrossRef] [Green Version]
  108. Yadav, A.; Kumar, B.; Datta, J.; Teknos, T.N.; Kumar, P. IL-6 Promotes Head and Neck Tumor Metastasis by Inducing Epithelial-Mesenchymal Transition via the JAK-STAT3-SNAIL Signaling Pathway. Mol. Cancer Res. MCR 2011, 9, 1658–1667. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Visciano, C.; Liotti, F.; Prevete, N.; Cali’, G.; Franco, R.; Collina, F.; de Paulis, A.; Marone, G.; Santoro, M.; Melillo, R.M. Mast Cells Induce Epithelial-to-Mesenchymal Transition and Stem Cell Features in Human Thyroid Cancer Cells through an IL-8-Akt-Slug Pathway. Oncogene 2015, 34, 5175–5186. [Google Scholar] [CrossRef] [PubMed]
  110. Lee, C.-H.; Chang, J.S.-M.; Syu, S.-H.; Wong, T.-S.; Chan, J.Y.-W.; Tang, Y.-C.; Yang, Z.-P.; Yang, W.-C.; Chen, C.-T.; Lu, S.-C.; et al. IL-1β Promotes Malignant Transformation and Tumor Aggressiveness in Oral Cancer. J. Cell. Physiol. 2015, 230, 875–884. [Google Scholar] [CrossRef] [PubMed]
  111. Lee, S.Y.; Ju, M.K.; Jeon, H.M.; Lee, Y.J.; Kim, C.H.; Park, H.G.; Han, S.I.; Kang, H.S. Reactive Oxygen Species Induce Epithelial-mesenchymal Transition, Glycolytic Switch, and Mitochondrial Repression through the Dlx-2/Snail Signaling Pathways in MCF-7 Cells. Mol. Med. Rep. 2019, 20, 2339–2346. [Google Scholar] [CrossRef] [PubMed]
  112. McMahon, G. VEGF Receptor Signaling in Tumor Angiogenesis. Oncologist 2000, 5, 3–10. [Google Scholar] [CrossRef] [PubMed]
  113. Feurino, L.W.; Zhang, Y.; Bharadwaj, U.; Zhang, R.; Li, F.; Fisher, W.E.; Brunicardi, F.C.; Chen, C.; Yao, Q.; Min, L. IL-6 Stimulates Th2 Type Cytokine Secretion and Upregulates VEGF and NRP-1 Expression in Pancreatic Cancer Cells. Cancer Biol. Ther. 2007, 6, 1096–1100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Boreddy, S.R.; Sahu, R.P.; Srivastava, S.K. Benzyl Isothiocyanate Suppresses Pancreatic Tumor Angiogenesis and Invasion by Inhibiting HIF-α/VEGF/Rho-GTPases: Pivotal Role of STAT-3. PLoS ONE 2011, 6, e25799. [Google Scholar] [CrossRef]
  115. Leibovich, S.J.; Polverini, P.J.; Shepard, H.M.; Wiseman, D.M.; Shively, V.; Nuseir, N. Macrophage-Induced Angiogenesis Is Mediated by Tumour Necrosis Factor-α. Nature 1987, 329, 630–632. [Google Scholar] [CrossRef]
  116. Gagari, E.; Tsai, M.; Lantz, C.S.; Fox, L.G.; Galli, S.J. Differential Release of Mast Cell Interleukin-6 Via c-Kit. Blood 1997, 89, 2654–2663. [Google Scholar] [CrossRef]
  117. Huang, M.; Pang, X.; Karalis, K.; Theoharides, T.C. Stress-Induced Interleukin-6 Release in Mice Is Mast Cell-Dependent and More Pronounced in Apolipoprotein E Knockout Mice. Cardiovasc. Res. 2003, 59, 241–249. [Google Scholar] [CrossRef] [Green Version]
  118. Gaje, P.N.; Amalia Ceausu, R.; Jitariu, A.; Stratul, S.I.; Rusu, L.-C.; Popovici, R.A.; Raica, M. Mast Cells: Key Players in the Shadow in Oral Inflammation and in Squamous Cell Carcinoma of the Oral Cavity. BioMed Res. Int. 2016, 2016, 9235080. [Google Scholar] [CrossRef] [Green Version]
  119. Nigrovic, P.A.; Binstadt, B.A.; Monach, P.A.; Johnsen, A.; Gurish, M.; Iwakura, Y.; Benoist, C.; Mathis, D.; Lee, D.M. Mast Cells Contribute to Initiation of Autoantibody-Mediated Arthritis via IL-1. Proc. Natl. Acad. Sci. USA 2007, 104, 2325–2330. [Google Scholar] [CrossRef]
  120. Hu, Z.Q.; Kobayashi, K.; Zenda, N.; Shimamura, T. Tumor Necrosis Factor-Alpha- and Interleukin-6-Triggered Mast Cell Development from Mouse Spleen Cells. Blood 1997, 89, 526–533. [Google Scholar] [CrossRef]
  121. Yanagida, M.; Fukamachi, H.; Ohgami, K.; Kuwaki, T.; Ishii, H.; Uzumaki, H.; Amano, K.; Tokiwa, T.; Mitsui, H.; Saito, H.; et al. Effects of T-Helper 2-Type Cytokines, Interleukin-3 (IL-3), IL-4, IL-5, and IL-6 on the Survival of Cultured Human Mast Cells. Blood 1995, 86, 3705–3714. [Google Scholar] [CrossRef] [PubMed]
  122. Cruse, G.; Cockerill, S.; Bradding, P. IgE Alone Promotes Human Lung Mast Cell Survival through the Autocrine Production of IL-6. BMC Immunol. 2008, 9, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Gulliksson, M.; Carvalho, R.F.S.; Ullerås, E.; Nilsson, G. Mast Cell Survival and Mediator Secretion in Response to Hypoxia. PLoS ONE 2010, 5, e12360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Hültner, L.; Kölsch, S.; Stassen, M.; Kaspers, U.; Kremer, J.-P.; Mailhammer, R.; Moeller, J.; Broszeit, H.; Schmitt, E. In Activated Mast Cells, IL-1 Up-Regulates the Production of Several Th2-Related Cytokines Including IL-9. J. Immunol. 2000, 164, 5556–5563. [Google Scholar] [CrossRef]
  125. Marcenaro, S.; Gallo, F.; Martini, S.; Santoro, A.; Griffiths, G.M.; Aricó, M.; Moretta, L.; Pende, D. Analysis of Natural Killer–Cell Function in Familial Hemophagocytic Lymphohistiocytosis (FHL): Defective CD107a Surface Expression Heralds Munc13-4 Defect and Discriminates between Genetic Subtypes of the Disease. Blood 2006, 108, 2316–2323. [Google Scholar] [CrossRef] [Green Version]
  126. Villanueva, J.; Lee, S.; Giannini, E.H.; Graham, T.B.; Passo, M.H.; Filipovich, A.; Grom, A.A. Natural Killer Cell Dysfunction Is a Distinguishing Feature of Systemic Onset Juvenile Rheumatoid Arthritis and Macrophage Activation Syndrome. Arthritis Res. Ther. 2004, 7, R30. [Google Scholar] [CrossRef] [Green Version]
  127. Lee, D.W.; Gardner, R.; Porter, D.L.; Louis, C.U.; Ahmed, N.; Jensen, M.; Grupp, S.A.; Mackall, C.L. Current Concepts in the Diagnosis and Management of Cytokine Release Syndrome. Blood 2014, 124, 188–195. [Google Scholar] [CrossRef] [Green Version]
  128. Cifaldi, L.; Prencipe, G.; Caiello, I.; Bracaglia, C.; Locatelli, F.; De Benedetti, F.; Strippoli, R. Inhibition of Natural Killer Cell Cytotoxicity by Interleukin-6: Implications for the Pathogenesis of Macrophage Activation Syndrome. Arthritis Rheumatol. 2015, 67, 3037–3046. [Google Scholar] [CrossRef]
  129. Wiley, S.R.; Schooley, K.; Smolak, P.J.; Din, W.S.; Huang, C.P.; Nicholl, J.K.; Sutherland, G.R.; Smith, T.D.; Rauch, C.; Smith, C.A. Identification and Characterization of a New Member of the TNF Family That Induces Apoptosis. Immunity 1995, 3, 673–682. [Google Scholar] [CrossRef] [Green Version]
  130. Prager, I.; Watzl, C. Mechanisms of Natural Killer Cell-Mediated Cellular Cytotoxicity. J. Leukoc. Biol. 2019, 105, 1319–1329. [Google Scholar] [CrossRef]
  131. Zamai, L.; Ahmad, M.; Bennett, I.M.; Azzoni, L.; Alnemri, E.S.; Perussia, B. Natural Killer (NK) Cell–Mediated Cytotoxicity: Differential Use of  TRAIL and Fas Ligand by Immature and Mature Primary Human NK Cells. J. Exp. Med. 1998, 188, 2375–2380. [Google Scholar] [CrossRef] [PubMed]
  132. Jewett, A.; Cavalcanti, M.; Bonavida, B. Pivotal Role of Endogenous TNF-Alpha in the Induction of Functional Inactivation and Apoptosis in NK Cells. J. Immunol. 1997, 159, 4815–4822. [Google Scholar] [CrossRef]
  133. Jin, F.; Wu, Z.; Hu, X.; Zhang, J.; Gao, Z.; Han, X.; Qin, J.; Li, C.; Wang, Y. The PI3K/Akt/GSK-3β/ROS/EIF2B Pathway Promotes Breast Cancer Growth and Metastasis via Suppression of NK Cell Cytotoxicity and Tumor Cell Susceptibility. Cancer Biol. Med. 2019, 16, 38–54. [Google Scholar] [CrossRef] [Green Version]
  134. Romero, A.I.; Thorén, F.B.; Brune, M.; Hellstrand, K. NKp46 and NKG2D Receptor Expression in NK Cells with CD56dim and CD56bright Phenotype: Regulation by Histamine and Reactive Oxygen Species. Br. J. Haematol. 2006, 132, 91–98. [Google Scholar] [CrossRef] [PubMed]
  135. Myers, T.J.; Brennaman, L.H.; Stevenson, M.; Higashiyama, S.; Russell, W.E.; Lee, D.C.; Sunnarborg, S.W. Mitochondrial Reactive Oxygen Species Mediate GPCR–Induced TACE/ADAM17-Dependent Transforming Growth Factor-α Shedding. Mol. Biol. Cell 2009, 20, 5236–5249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Brill, A.; Chauhan, A.K.; Canault, M.; Walsh, M.T.; Bergmeier, W.; Wagner, D.D. Oxidative Stress Activates ADAM17/TACE and Induces Its Target Receptor Shedding in Platelets in a P38-Dependent Fashion. Cardiovasc. Res. 2009, 84, 137–144. [Google Scholar] [CrossRef] [Green Version]
  137. Wiernik, A.; Foley, B.; Zhang, B.; Verneris, M.R.; Warlick, E.; Gleason, M.K.; Ross, J.A.; Luo, X.; Weisdorf, D.J.; Walcheck, B.; et al. Targeting Natural Killer Cells to Acute Myeloid Leukemia In Vitro with a CD16 × 33 Bispecific Killer Cell Engager and ADAM17 Inhibition. Clin. Cancer Res. 2013, 19, 3844–3855. [Google Scholar] [CrossRef] [Green Version]
  138. Tofaris, G.K.; Patterson, P.H.; Jessen, K.R.; Mirsky, R. Denervated Schwann Cells Attract Macrophages by Secretion of Leukemia Inhibitory Factor (LIF) and Monocyte Chemoattractant Protein-1 in a Process Regulated by Interleukin-6 and LIF. J. Neurosci. Off. J. Soc. Neurosci. 2002, 22, 6696–6703. [Google Scholar] [CrossRef]
  139. Austin, P.J.; Moalem-Taylor, G. The Neuro-Immune Balance in Neuropathic Pain: Involvement of Inflammatory Immune Cells, Immune-like Glial Cells and Cytokines. J. Neuroimmunol. 2010, 229, 26–50. [Google Scholar] [CrossRef]
  140. Whitehead, K.J.; Smith, C.G.S.; Delaney, S.-A.; Curnow, S.J.; Salmon, M.; Hughes, J.P.; Chessell, I.P. Dynamic Regulation of Spinal Pro-Inflammatory Cytokine Release in the Rat in Vivo Following Peripheral Nerve Injury. Brain. Behav. Immun. 2010, 24, 569–576. [Google Scholar] [CrossRef]
  141. Schäfers, M.; Sorkin, L. Effect of Cytokines on Neuronal Excitability. Neurosci. Lett. 2008, 437, 188–193. [Google Scholar] [CrossRef]
  142. Pollock, J.; McFarlane, S.M.; Connell, M.C.; Zehavi, U.; Vandenabeele, P.; MacEwan, D.J.; Scott, R.H. TNF-α Receptors Simultaneously Activate Ca2+ Mobilisation and Stress Kinases in Cultured Sensory Neurones. Neuropharmacology 2002, 42, 93–106. [Google Scholar] [CrossRef] [PubMed]
  143. Junger, H.; Sorkin, L.S. Nociceptive and Inflammatory Effects of Subcutaneous TNFα. Pain 2000, 85, 145–151. [Google Scholar] [CrossRef] [PubMed]
  144. Fukuoka, H.; Kawatani, M.; Hisamitsu, T.; Takeshige, C. Cutaneous Hyperalgesia Induced by Peripheral Injection of Interleukin-1β in the Rat. Brain Res. 1994, 657, 133–140. [Google Scholar] [CrossRef] [PubMed]
  145. Wolf, G.; Gabay, E.; Tal, M.; Yirmiya, R.; Shavit, Y. Genetic Impairment of Interleukin-1 Signaling Attenuates Neuropathic Pain, Autotomy, and Spontaneous Ectopic Neuronal Activity, Following Nerve Injury in Mice. Pain 2006, 120, 315–324. [Google Scholar] [CrossRef] [PubMed]
  146. Schäfers, M.; Brinkhoff, J.; Neukirchen, S.; Marziniak, M.; Sommer, C. Combined Epineurial Therapy with Neutralizing Antibodies to Tumor Necrosis Factor-Alpha and Interleukin-1 Receptor Has an Additive Effect in Reducing Neuropathic Pain in Mice. Neurosci. Lett. 2001, 310, 113–116. [Google Scholar] [CrossRef]
  147. Tobinick, E.; Davoodifar, S. Efficacy of Etanercept Delivered by Perispinal Administration for Chronic Back and/or Neck Disc-Related Pain: A Study of Clinical Observations in 143 Patients. Curr. Med. Res. Opin. 2004, 20, 1075–1085. [Google Scholar] [CrossRef]
  148. Cohen, S.P.; Bogduk, N.; Dragovich, A.; Buckenmaier, C.C.; Griffith, S.; Kurihara, C.; Raymond, J.; Richter, P.J.; Williams, N.; Yaksh, T.L. Randomized, Double-Blind, Placebo-Controlled, Dose-Response, and Preclinical Safety Study of Transforaminal Epidural Etanercept for the Treatment of Sciatica. Anesthesiology 2009, 110, 1116–1126. [Google Scholar] [CrossRef] [Green Version]
  149. Ohtori, S.; Miyagi, M.; Eguchi, Y.; Inoue, G.; Orita, S.; Ochiai, N.; Kishida, S.; Kuniyoshi, K.; Nakamura, J.; Aoki, Y.; et al. Efficacy of Epidural Administration of Anti-Interleukin-6 Receptor Antibody onto Spinal Nerve for Treatment of Sciatica. Eur. Spine J. 2012, 21, 2079–2084. [Google Scholar] [CrossRef] [Green Version]
  150. Szatrowski, T.P.; Nathan, C.F. Production of Large Amounts of Hydrogen Peroxide by Human Tumor Cells. Cancer Res. 1991, 51, 794–798. [Google Scholar]
  151. Weinberg, F.; Ramnath, N.; Nagrath, D. Reactive Oxygen Species in the Tumor Microenvironment: An Overview. Cancers 2019, 11, 1191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Conklin, K.A. Chemotherapy-Associated Oxidative Stress: Impact on Chemotherapeutic Effectiveness. Integr. Cancer Ther. 2004, 3, 294–300. [Google Scholar] [CrossRef] [PubMed]
  153. Areti, A.; Yerra, V.G.; Naidu, V.; Kumar, A. Oxidative Stress and Nerve Damage: Role in Chemotherapy Induced Peripheral Neuropathy. Redox Biol. 2014, 2, 289–295. [Google Scholar] [CrossRef] [Green Version]
  154. Park, E.-S.; Gao, X.; Chung, J.M.; Chung, K. Levels of Mitochondrial Reactive Oxygen Species Increase in Rat Neuropathic Spinal Dorsal Horn Neurons. Neurosci. Lett. 2006, 391, 108–111. [Google Scholar] [CrossRef]
  155. Schwartz, E.S.; Lee, I.; Chung, K.; Mo Chung, J. Oxidative Stress in the Spinal Cord Is an Important Contributor in Capsaicin-Induced Mechanical Secondary Hyperalgesia in Mice. Pain 2008, 138, 514–524. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Schwartz, E.S.; Kim, H.Y.; Wang, J.; Lee, I.; Klann, E.; Chung, J.M.; Chung, K. Persistent Pain Is Dependent on Spinal Mitochondrial Antioxidant Levels. J. Neurosci. 2009, 29, 159–168. [Google Scholar] [CrossRef] [Green Version]
  157. Fidanboylu, M.; Griffiths, L.A.; Flatters, S.J.L. Global Inhibition of Reactive Oxygen Species (ROS) Inhibits Paclitaxel-Induced Painful Peripheral Neuropathy. PLoS ONE 2011, 6, e25212. [Google Scholar] [CrossRef]
  158. Soriani, A.; Iannitto, M.L.; Ricci, B.; Fionda, C.; Malgarini, G.; Morrone, S.; Peruzzi, G.; Ricciardi, M.R.; Petrucci, M.T.; Cippitelli, M.; et al. Reactive Oxygen Species– and DNA Damage Response–Dependent NK Cell Activating Ligand Upregulation Occurs at Transcriptional Levels and Requires the Transcriptional Factor E2F1. J. Immunol. 2014, 193, 950–960. [Google Scholar] [CrossRef] [Green Version]
  159. Ledeboer, A.; Jekich, B.M.; Sloane, E.M.; Mahoney, J.H.; Langer, S.J.; Milligan, E.D.; Martin, D.; Maier, S.F.; Johnson, K.W.; Leinwand, L.A.; et al. Intrathecal Interleukin-10 Gene Therapy Attenuates Paclitaxel-Induced Mechanical Allodynia and Proinflammatory Cytokine Expression in Dorsal Root Ganglia in Rats. Brain. Behav. Immun. 2007, 21, 686–698. [Google Scholar] [CrossRef] [Green Version]
  160. Echeverry, S.; Shi, X.Q.; Haw, A.; Liu, G.; Zhang, Z.; Zhang, J. Transforming Growth Factor-Β1 Impairs Neuropathic Pain through Pleiotropic Effects. Mol. Pain 2009, 5, 1744-8069-5-16. [Google Scholar] [CrossRef] [Green Version]
Figure 1. The potential role of mast cells (MCs) and NK cells in the pathobiology of CIPN. (A). Intact neuron prior to insults by mast cells (B). Mast cells induce neuroinflammation by MC release of mediators and neuropeptides, which in turn mediate releases of algogenic substances from neuronal endings. (C). 161533 TriKE augments ADCC of NK cells against CD33 expressing MCs. (D). NK cells release cytotoxic granules, which results in complete loss of neuropathic nerve fibers, which can mitigate neuropathic symptoms.
Figure 1. The potential role of mast cells (MCs) and NK cells in the pathobiology of CIPN. (A). Intact neuron prior to insults by mast cells (B). Mast cells induce neuroinflammation by MC release of mediators and neuropeptides, which in turn mediate releases of algogenic substances from neuronal endings. (C). 161533 TriKE augments ADCC of NK cells against CD33 expressing MCs. (D). NK cells release cytotoxic granules, which results in complete loss of neuropathic nerve fibers, which can mitigate neuropathic symptoms.
Ijms 24 12543 g001
Figure 2. The impact of proinflammatory cytokines on chemotherapy-induced peripheral neuropathy (CIPN). Proinflammatory cytokines can exert negative multifaceted effects in CIPN by disease progression of cancer, activation and maintenance of mast cells (MCs), NK cell dysfunction, neuroinflammation and nociceptor sensitization.
Figure 2. The impact of proinflammatory cytokines on chemotherapy-induced peripheral neuropathy (CIPN). Proinflammatory cytokines can exert negative multifaceted effects in CIPN by disease progression of cancer, activation and maintenance of mast cells (MCs), NK cell dysfunction, neuroinflammation and nociceptor sensitization.
Ijms 24 12543 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Yun, H.D.; Goel, Y.; Gupta, K. Crosstalk of Mast Cells and Natural Killer Cells with Neurons in Chemotherapy-Induced Peripheral Neuropathy. Int. J. Mol. Sci. 2023, 24, 12543. https://doi.org/10.3390/ijms241612543

AMA Style

Yun HD, Goel Y, Gupta K. Crosstalk of Mast Cells and Natural Killer Cells with Neurons in Chemotherapy-Induced Peripheral Neuropathy. International Journal of Molecular Sciences. 2023; 24(16):12543. https://doi.org/10.3390/ijms241612543

Chicago/Turabian Style

Yun, Hyun Don, Yugal Goel, and Kalpna Gupta. 2023. "Crosstalk of Mast Cells and Natural Killer Cells with Neurons in Chemotherapy-Induced Peripheral Neuropathy" International Journal of Molecular Sciences 24, no. 16: 12543. https://doi.org/10.3390/ijms241612543

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