Next Article in Journal
Marine Bioactive Peptides: Anti-Photoaging Mechanisms and Potential Skin Protective Effects
Next Article in Special Issue
An Elevated IL10 mRNA Combined with Lower TNFA mRNA Level in Active Rheumatoid Arthritis Peripheral Blood
Previous Article in Journal
Ghrelin in Focus: Dissecting Its Critical Roles in Gastrointestinal Pathologies and Therapies
Previous Article in Special Issue
A Potential Role for the Receptor for Advanced Glycation End-Products (RAGE) in the Development of Secondhand Smoke-Induced Chronic Sinusitis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Nerve Growth Factor and the Role of Inflammation in Tumor Development

by
Giampiero Ferraguti
1,†,
Sergio Terracina
1,†,
Luigi Tarani
2,
Francesca Fanfarillo
1,
Sara Allushi
1,
Brunella Caronti
3,
Paola Tirassa
4,
Antonella Polimeni
5,
Marco Lucarelli
1,6,
Luca Cavalcanti
7,
Antonio Greco
7 and
Marco Fiore
4,*
1
Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
2
Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, 00185 Rome, Italy
3
Department of Human Neurosciences, Sapienza University Hospital of Rome, 00185 Rome, Italy
4
Institute of Biochemistry and Cell Biology (IBBC-CNR), Department of Sensory Organs, Sapienza University of Rome, 00185 Rome, Italy
5
Department of Odontostomatological and Maxillofacial Sciences, Sapienza University of Rome, 00185 Rome, Italy
6
Pasteur Institute, Cenci Bolognetti Foundation, Sapienza University of Rome, 00185 Rome, Italy
7
Department of Sensory Organs, Sapienza University of Rome, 00185 Rome, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Curr. Issues Mol. Biol. 2024, 46(2), 965-989; https://doi.org/10.3390/cimb46020062
Submission received: 6 December 2023 / Revised: 12 January 2024 / Accepted: 19 January 2024 / Published: 23 January 2024
(This article belongs to the Special Issue Advances in Understanding Molecular Basis of Inflammatory Diseases)

Abstract

:
Nerve growth factor (NGF) plays a dual role both in inflammatory states and cancer, acting both as a pro-inflammatory and oncogenic factor and as an anti-inflammatory and pro-apoptotic mediator in a context-dependent way based on the signaling networks and its interaction with diverse cellular components within the microenvironment. This report aims to provide a summary and subsequent review of the literature on the role of NGF in regulating the inflammatory microenvironment and tumor cell growth, survival, and death. The role of NGF in inflammation and tumorigenesis as a component of the inflammatory system, its interaction with the various components of the respective microenvironments, its ability to cause epigenetic changes, and its role in the treatment of cancer have been highlighted in this paper.

1. Introduction

Inflammatory processes play a multifaceted role in tumor development and progression [1,2,3,4,5,6,7]. While the immune system’s inflammatory response is typically a defense mechanism against infections and tissue damage, chronic or persistent inflammation can contribute to the initiation, growth, and spread of certain types of tumors. Thus, the understanding of the relationship between inflammation and cancer could better focus on anti-inflammatory therapies and drugs that target inflammatory pathways as potential strategies for cancer prevention and treatment [8]. In recent years, a growing body of evidence has emphasized the involvement of neurotrophins (NTs) in the complex landscape of both inflammation and cancer biology, showing their significant role in determining tumor cell growth and survival, particularly in certain types of cancers expressing NT receptors on their cell surfaces [9,10]. One of the most well-studied NTs is the nerve growth factor (NGF), which binds to its specific receptor, tropomyosin-related kinase A (TrkA), expressed on various types of cancer cells, including those derived from the brain, prostate, breast, and pancreas, among others [11,12,13]. NGF contributes to inflammation by acting as a signaling molecule that stimulates immune cells to release cytokines and enhances the sensitivity of sensory nerves, contributing to the perception of pain and hypersensitivity in inflamed tissues [14]. NTs promote tumor growth by stimulating cancer cell proliferation and suppressing apoptosis, which physiologically helps eliminate damaged or unwanted cells from the body [15]. Furthermore, NTs can stimulate the production of pro-angiogenic factors, modulate the tumor microenvironment (TME), and induce the epithelial–mesenchymal transition (EMT), leading to increased cell mobility and invasiveness [16].
Overall, the activation of NGF signaling pathways in cancer cells can contribute to aggressive tumor growth, metastasis, and therapy resistance. Originally identified for its pivotal role in neuronal development and function, NGF has emerged as a critical player in the growth and survival of various tumor types. The intriguing aspect of NGF’s contribution to cancer lies in its dual role, acting as both an oncogenic factor, fueling tumor cell growth, and as a pro-apoptotic mediator, promoting tumor cell death under certain circumstances [17,18,19]. The context-dependent actions of NGF in cancer underscore the complexity of the signaling networks in which it operates and its interaction with diverse cellular components within the TME. Overall, NGF’s involvement in both inflammation and tumors further highlights its complex role in regulating physiological processes and disease states. Understanding the intricate interactions of NGF with various cell types and pathways is crucial for developing targeted therapies that can modulate its effects in different disease contexts, offering a potential therapeutic strategy to inhibit tumor progression and improve patient outcomes. However, further research is needed to fully understand the complexities of NGF signaling in different cancer types and to develop effective and safe targeted therapies.
This narrative report aims to provide a summary and subsequent review of literature evidence on the role of NGF and its role in regulating tumor cell growth and death as an inflammatory factor.

2. NGF and Inflammation and Tumor Growth

2.1. Nerve Growth Factor and Neurotrophins

NGF is a crucial neurotrophic factor responsible for the growth, survival, and maintenance of neuronal and non-neuronal cells [20,21]. Discovered in the 1950s, it was one of the first NTs identified and extensively studied for its role in the development and function of the nervous system [22,23]. NGF belongs to the family of NTs, which includes other proteins like Brain-Derived Neurotrophic Factor (BDNF), Neurotrophin-3 (NT-3), and Neurotrophin-4/5 (NT-4/5) [24,25]. NGF is synthesized as a precursor, called pro-NGF, which after processing generates the mature NGF molecule. NGF is primarily produced by various cell types, including immune cells, endothelial cells, and tissues within the nervous system [14,26,27,28].
Pro-NGF has a physiological role that goes beyond that of a simple precursor, especially in the nervous system, where it may possess pro-apoptotic activity [29]. Indeed, pro-NGF regulates apoptosis and inflammation and is associated with several neurodegenerative diseases, myocardial infarction, and diabetes [30,31,32,33,34]. The actions of NGF are mediated through its interaction with specific receptors. There are two primary receptors associated with NGF:
(1)
TrkA is the high-affinity receptor for NGF; its activation triggers a cascade of intracellular signaling events, including the MAPK/ERK pathway and the PI3K/Akt pathway [35,36]. These pathways play essential roles in cell growth, differentiation, and survival. TrkA is expressed on the surface of neurons and other cell types, enabling NGF to exert its neurotrophic effects;
(2)
p75 neurotrophin receptor (p75NTR) has a lower affinity for NGF but plays a modulatory role in NGF signaling [20,37]. It can interact with TrkA and enhance its binding affinity for NGF, influencing the cellular responses elicited by NGF. p75NTR is mostly involved in processes like cell death, survival decisions, and axonal growth.
Furthermore, NGF may attach to the membrane receptor sortilin, which has been demonstrated to participate in cancer growth [38], and also to neuropilin-1 (NRP1), a nociceptor-enriched co-receptor for NGF that is necessary for the TrkA signaling of pain [39,40,41].
Indeed, modifications in NGF levels in the serum and plasma have been shown during the beginning and evolution of many health conditions, including the post-partum period, stressful events, cardiometabolic disruptions, aging, alcohol addiction, and other pathophysiological conditions, such as psychiatric, neurological, and immune disorders [42,43,44,45,46,47,48,49,50,51,52]. Thus, the understanding of the interplay between NGF and its receptors is crucial not only for comprehending the complexities of neuronal development and function but also for exploring potential therapeutic interventions in various diseases, including neurodegenerative disorders and certain types of cancers where NGF signaling may be implicated [51,53,54,55].

2.2. Inflammation

Inflammation is a natural response by the body’s immune system to protect against harmful stimuli like pathogens, irritants, or damaged cells. It is a complex biological process that aims to eliminate the initial cause of cell injury, clear out damaged cells, and initiate tissue repair [56]. Actually, in medicine, inflammation, a term coined by the Romans, lacks a precise, universally accepted definition, varying in interpretation based on context and individual perspectives [57]. It often carries a negative connotation as an uncontrolled reaction likened to a destructive wildfire, requiring immediate containment.
However, overshadowed in this view is the fundamental role that inflammation plays in both maintaining health and ensuring survival. Inflammation involves a complex interplay of cells, chemicals, and molecular signals including blood vessel dilatation (causing redness and heat) and increased permeability, allowing immune cells and fluids to move from the bloodstream into the tissues (leading to swelling); the cellular release of chemicals such as histamine, cytokines, and prostaglandins, which help to trigger the immune response and promote healing; and finally the activation of immune cells which migrate to the affected area to destroy pathogens or damaged cells [58].
Cytokines play a pivotal role in orchestrating or modulating inflammation, confirming not only the presence and magnitude of inflammation but also guiding treatment decisions. Inflammation can be distinguished as acute and chronic. Acute inflammation is the body’s immediate and short-term response to an injury or infection [59]. It is characterized by symptoms like redness, swelling, heat, pain, and sometimes loss of function [60,61]. Chronic inflammation, on the other hand, is long-term and can last for weeks, months, or even years [62]. It occurs when the immune system’s response persists, often due to underlying health conditions, such as autoimmune disorders, ongoing infections, obesity, or prolonged exposure to irritants like smoke.
Lifestyle factors like stress, poor diet, lack of exercise, and environmental toxins can contribute to chronic inflammation [63]. While inflammation is a vital part of the body’s defense mechanism, chronic inflammation can be a problematic event leading to tissue damage and various diseases including diabetes, cancer, cardiovascular diseases, eye disorders, arthritis, obesity, autoimmune diseases, and inflammatory bowel disease [64].
Interestingly, inflammation is at the base of various conditions and a plethora of etiopathogenetic events; for example, hepatitis (liver inflammation) has been associated with viral infections, excessive alcohol consumption, certain medications, or autoimmune responses [62,65,66,67,68]. Long-term inflammation of the liver can lead to cirrhosis and liver cancer [69,70]. Inflammatory states and diseases cover a wide range of conditions affecting various parts of the body, varying in severity and requiring different treatments, including medications, lifestyle changes, and sometimes, in more severe cases, surgical interventions or specialized therapies.
The widespread use of anti-inflammatory medications assumed to counteract all inflammatory responses potentially may hinder the body’s ability to fully recover [71,72]. Indeed, not all situations warrant an inflammatory response (such as blunt trauma and exposure to toxins), but since inflammation affects both unhealthy and healthy tissues without discrimination, it should be treated when it has the potential to persist or spread uncontrollably, causing prolonged damage. Effective management often entangles a multidisciplinary approach that requires the support of specialized healthcare professionals in this specific condition. Actually, it has been suggested that an effective way to guide therapy for inflammation is to assess a combination of markers associated with inflammation and fibrosis, such as C-reactive protein, ferritin, serum amyloid A (SAA), pro-calcitonin, and transforming growth factor-β (TGF-β, a significant contributor to fibrosis), alongside cytokine profiling [73].

2.3. Role of NGF in Carcinogenesis

NGF plays a significant role in various aspects of human health, including its involvement in tumors. Overall, evidence indicates that NGF is unable to generate cell carcinogenesis alone, both in normal neuronal and non-neuronal cells/tissues; however, it could be a major determinant in the case of co-expression with pro-carcinogenic molecules [74]. Quite intriguingly, NGF was initially discovered by R. Levi-Montalcini nearly 60 years ago in the context of a transplantation experiment involving a malignant mouse sarcoma [75,76].

NGF as a Tumor Growth Facilitator or Suppressor

Depending on the tumor’s origin, pro-survival signaling can be facilitated through TrkA and/or p75NTR receptors [77]. In breast cancer, NGF plays a crucial role in stimulating proliferative signaling via TrkA and pro-survival signaling through p75NTR [35]. Furthermore, the activation of p75NTR in breast cancer promotes increased resistance to cell death induced by chemotherapeutic treatments. On the other hand, the role of p75NTR in prostate cells is distinct since p75NTR mediates cell death and acts as a tumor suppressor in the case of normal prostate cells [78]. In prostate cancer, the expression of p75NTR is lost, contributing to tumor progression, death evasion, uncontrolled proliferation, and metastasis to distant sites [79]. Interestingly, other mechanisms were found in recent studies. For example, NGF plays a significant role in liver cancer progression and metastasis, exerting wide influences on liver cancer cell polarity and motility by regulating signaling pathways involved in cell movement, cytoskeletal organization, and cellular polarity [80]. Heightened NGF disrupts cell polarity, boosts cell movement, triggers changes related to cell transition, rearranges the cell’s structural framework, and protects cells from apoptosis and detachment-induced cell death [80]. In Table 1, we report the main role of NGF and its receptors in various cancers.
While the primary role of NGF is related to the development and function of nerve cells, it also plays a main role in inflammation. Inflammation, as stated before, is a complex biological response triggered by the body’s immune system to protect against harmful stimuli, such as tissue damage and pathogens. In this scenario, NGF can regulate the innervation and neuronal activity of peripheral neurons, inducing the release of immune-active cytokines, neuropeptides, and neurotransmitters [14,187,188,189]. Furthermore, NGF can also directly influence innate and adaptive immune responses through its interaction with various cells involved in the immune response, including mast cells, lymphocytes, and macrophages [28,190]. Actually, NGF has a variety of effects that can be either pro-inflammatory or anti-inflammatory depending on the expression of its receptors, which are dynamically regulated in immune cells depending on their state of differentiation and functional activity [191,192]. The seeming ambiguity is mainly due to the role of NGF as an endogenous molecule capable of triggering immune responses while also initiating pathways that control inflammation and prevent excessive tissue damage so that altered expression of its receptors could hinder NGF’s ability to engage the regulatory feedback processes for finally sustaining the perpetuation of inflammation in conditions such as chronic inflammatory diseases or autoimmune disorders [14]. Additionally, as NGF can contribute to the sensitivity and pain associated with the neurogenic inflammation of tissues, a potential role of NTs has been suggested as novel treatment strategies in chronic inflammatory diseases [193,194].
More specifically, NGF has intricate connections with neuroinflammation, which involves complex interactions between immune cells, glial cells, and various signaling molecules. Microglia act as the resident immune cells of the central nervous system (CNS) and can become activated in response to injury or inflammation [195]. NGF can modulate the activation and function of glial cells, particularly microglia and astrocytes, influencing their release of inflammatory mediators [196]. It can both promote and dampen the release of various cytokines depending on the context, contributing to the fine-tuning of the inflammatory response in the CNS [157,197,198] (see Figure 1).
On the other hand, NGF exhibits neuroprotective effects by mitigating the harmful consequences of neuroinflammation and supporting neuronal survival and function, potentially counteracting the detrimental effects of excessive inflammation on neurons [199,200]. Furthermore, dysregulation of NGF signaling has been associated with various neurological disorders characterized by neuroinflammation, such as Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, and neuropathic pain conditions [201,202]. In these disorders, altered NGF levels or signaling pathways contribute to the progression of neuroinflammation and neuronal damage, so it has been suggested that modulating NGF levels or its interactions within the CNS may offer potential avenues for managing neuroinflammatory conditions and related neurological disorders [203,204].
Unfortunately, inflammation plays a multifaceted role in tumor development and progression. While the immune system’s inflammatory response is typically a defense mechanism against infections and tissue damage, chronic or persistent inflammation can contribute to the initiation, growth, and spread of certain types of tumors in several ways [69]. Cycles of tissue damage and subsequent repair processes can create an environment conducive to genetic mutations and abnormalities, increasing the likelihood of cancerous changes in cells [205]. Inflammation can indeed sustain tissue damage and attract immune cells to the site of tissue damage.
Hallmarks of cancer-associated inflammation include the presence of infiltrating leukocytes, cytokines, chemokines, growth factors, lipid messengers, and matrix-degrading enzymes [206].
Some of these immune cells, like certain types of macrophages and lymphocytes, can produce factors that support tumor growth and suppress the immune system’s ability to eliminate cancer cells [207]. Inflammatory signals can stimulate angiogenesis for oxygen provision and nutrients to the tumor cells, aiding their proliferation and survival [10,208]. Managing chronic inflammation, either through lifestyle changes or medication, may play a role in reducing the risk of certain cancers or improving treatment outcomes [209].

3. NGF and the TME

The TME is a complex, highly heterogeneous, and dynamic ecosystem of cells, molecules, and structures that surround and interact with cancer cells within a tumor [210]. The TME is mainly constituted of cancer cells, stromal cells, immune cells, ECM, blood vessels, and signaling molecules [211]. It is often characterized by immune evasion, hypoxia, acidic pH, and stromal changes. The TME is significantly influenced by inflammation, creating a complex milieu that can either promote or inhibit tumor growth [212]. Chronic inflammation in the TME can create a supportive environment for tumor growth, survival, and metastasis by fostering angiogenesis, extracellular matrix (ECM) remodeling, and immune suppression [213]. Before metastasis occurs, the primary tumor can alter the TME within a particular tissue, leading to the initiation of a pre-metastatic niche (PMN) [214]. This phenomenon presents a valuable window of opportunity for the primary tumor cells to adapt, survive, and subsequently thrive, ultimately creating favorable conditions for colonization in the target organ during the subsequent metastatic process [215].
NGF can influence the TME, affecting the interactions between cancer cells and surrounding stromal cells, immune cells, and other components, contributing to both tumor growth and suppression [74]. On one hand, NGF can induce apoptosis in some cancer cells, limiting their survival, and may modulate immune responses within the TME, influencing the anti-tumor immune response [19]. One intriguing aspect of NGF’s protective function is its capacity to maintain the physiological activity within the tissue microenvironment, thereby safeguarding tissue and organ functionality. This effect has been notably observed in the preservation of corneal nerve cells and the regulation of homeostasis across various systems, including the nervous, immune, and endocrine systems [216]. On the other hand, evidence suggests that NGF can promote endothelial cell proliferation and angiogenesis by stimulating pro-angiogenic factors, facilitating the delivery of oxygen and nutrients to the tumor cells, and promoting tumor growth and metastasis [217]. Some of the most important pro-angiogenic factors activated by NGF in tumors include vascular endothelial growth factor (VEGF), which stimulates the growth and permeability of blood vessels, facilitating the supply of nutrients and oxygen to tumor cells; fibroblast growth factor (FGF), which plays a role in the proliferation and migration of endothelial cells, which form the inner lining of blood vessels; interleukin-8 (IL-8), a chemokine that can recruit and activate endothelial cells, promoting angiogenesis; and Matrix Metalloproteinases (MMPs), which are enzymes that degrade the ECM, allowing endothelial cells to migrate and form new blood vessels [140,218,219]. Furthermore, NGF can influence the stabilization and activation of hypoxia-inducible factor 1-alpha (HIF-1α), a transcription factor that plays a key role in regulating the expression of genes involved in angiogenesis in response to low oxygen levels (hypoxia) [100,158].
Two conceptual models, known as the “immunosurveillance model” and the “danger model,” present differing viewpoints regarding the interaction between the immune system and cancer cells [74,207]. The immunosurveillance model proposes that the immune system identifies tumor cells as foreign invaders and eradicates them. Conversely, the danger model suggests that immune cells do not recognize cancer cells as threats due to specific signaling deficits. In both cases, the composition of the TME emerges as a critical factor influencing these immune responses. The interaction between immune cells that infiltrate the tumor, those that reside within it, as well as epithelial and stromal cells, results in the release of a multitude of pro-inflammatory, anti-inflammatory, pro-angiogenic, and anti-angiogenic agents [220]. Significantly, NGF has been discovered to support the survival and functionality of diverse immune cell types, which encompass mast cells, lymphocytes, macrophages, granulocytes, T- and B-cell subtypes, natural killer cells, and eosinophils [52]. These immune cells not only exhibit responses to NGF but also can generate and release NGF themselves, implying a role for this molecule both in innate and adaptive immune responses [14].
In the context of the TME, increased levels of NGF may be released and engage with immune cells in autocrine or paracrine manners [221,222]. Increased NGF levels could contribute to the establishment of an immunosuppressive TME, thereby fostering resistance to immunotherapy. Tumors can contain nerve fibers that release NGF, which interacts with nerve endings present in the TME [223]. This crosstalk between nerves and the tumor can modulate immune responses [150]. In some cases, NGF may promote the expansion of regulatory T cells (Tregs), which can suppress the activity of cytotoxic T cells and dampen the anti-tumor immune response [224].
NGF can modulate the production of cytokines and chemokines in the TME, influencing the function of immune cells, creating an immunosuppressive milieu, and favoring tumor growth and immune escape. Furthermore, NGF may indirectly induce the expression of immune checkpoint molecules such as the programmed death-1 ligand (PD-L1), a co-inhibitory factor of the immune response, on tumor cells or immune cells within the TME, further inhibiting the activity of cytotoxic T cells [225]. NGF can influence the polarization of tumor-associated macrophages (TAMs) within the TME. TAMs can adopt different activation states, and some of these states can promote an immunosuppressive environment that hinders effective anti-tumor immunity [226]. Cancer cells utilize a mechanism involving the generation of reactive oxygen species (ROS) to activate c-Jun [227]. This activation prompts the secretion of NGF, which in turn primes nociceptive nerves to produce calcitonin gene-related peptide (CGRP). Subsequently, neurogenic CGRP induces a cellular self-protective process known as autophagy in cancer cells, achieved by disrupting the mTOR–Raptor interaction via Rap1 signaling. Interestingly, strategies aimed at starving cancer cells of nutrients by targeting glycolysis or angiogenesis can inadvertently exacerbate the reciprocal relationship between cancer cells and nociceptive nerves. To break this cycle and enhance therapeutic outcomes, it could be beneficial to intervene by inhibiting neurogenic CGRP.
NGF plays a central role in the intricate signaling network within Cancer Stem Cell (CSC) metabolism. CSCs are key elements of the TME, possessing the ability to self-renew and playing a pivotal role in the expansion and chronicity of malignant tumors favoring host immune surveillance escape, the release of pro-tumorigenic chemokines by innate immune cells, and the development of more aggressive tumors [9]. Some studies have suggested that NGF can activate signaling pathways that are associated with stem cell characteristics, such as the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway [141]. Activation of these pathways can contribute to the self-renewal and survival of CSCs [228]. NGF can affect the behavior of various cells within the TME, including immune cells, fibroblasts, and endothelial cells within the tumor, which can create an environment that supports CSC survival and function. The expansion and dissemination of CSCs are intricately linked to their energy supply, primarily through the insulin-like growth factor (IGF-1) receptor pathway controlling glucose uptake by acting on the insulin receptor substrate 1 (IRS1)/PI3K/AKT pathway and the nuclear factor kappa B (NF-Kb)/Snail/zinc finger E-box-binding homeobox 1 (ZEB1) pathway (crucial for the EMT) [229,230,231]. So, the insulin/IGF pathway collaborates with signaling platforms associated with cell proliferation and pluripotency, fostering the transcription of EMT factors. Now, NGF steps into the spotlight. The EMT is a biological process during which epithelial cells transform, losing their characteristic features and adopting mesenchymal properties. This transition results in heightened cell mobility and invasiveness, ultimately facilitating the spread of cancer cells to nearby tissues and distant organs. Emerging evidence suggests that NGF plays a significant role in triggering the EMT in specific cancer cell types, further enhancing their invasive potential and metastatic capabilities [232,233]. Furthermore, NGF has been shown to regulate insulin signaling and combat insulin resistance by enhancing glucose uptake, particularly in degenerating neurons, while TrkA can transactivate the insulin receptor (IR) signaling pathway and the oncogenic fusion protein Trk-T1, originating from thyroid carcinoma, can trigger the formation of the IRS–Growth factor receptor-bound protein 2 (IRS-Grb2) complex with the related pro-oncogenic processes (transformation of fibroblasts and intestinal epithelial cells). NGF signaling pathways have been shown to induce changes in cell morphology and enhance the migratory and invasive properties of these cancer cells, contributing to the aggressive behavior of tumors [80]. CSCs are often resistant to conventional treatments like chemotherapy and radiation, and NGF-mediated signaling pathways may enhance their ability to withstand these therapies. Given the potential role of NGF in CSC behavior, it has been suggested as a potential therapeutic target so that modulating NGF signaling pathways could be explored as a strategy to target CSCs and improve the effectiveness of cancer treatments.

4. NGF Receptor Expression in Cancer

Both NGF high-affinity receptors (Trk) and the low-affinity p75NTR receptor and their cross-talk have been found to play significant roles in various types of cancer (Table 1).
The activation of Trk receptors is initiated by the autophosphorylation of tyrosine residues within their intracellular domains. This activation subsequently triggers multiple downstream signaling pathways involving various enzymes and adaptor proteins, including extracellularly regulated kinases (ERKs), phosphatidylinositol 3-kinase (PI3K), and phospholipase C (PLC). These pathways collectively govern cell proliferation, differentiation, and survival, impacting both neuronal and non-neuronal cell types. Conversely, signaling through p75NTR leads to the activation of a c-jun N-terminal kinase (JNK) cascade, which is contingent on the specific adaptor proteins bound to the receptor. The activation of p75NTR, via JNK, leads to the activation of p53 and the expression of pro-apoptotic genes such as Bcl-2. Additionally, when the p75NTR collaborates with tumor necrosis factor receptor-associated factor 6 (TRAF6), it promotes the activation of the nuclear factor-kappa B (NF-kB) signaling pathway, which has a pro-survival effect. The diversity in activation signaling pathways mediated by NT receptors reflects the complexity of their functions, both within and outside the central nervous system, as well as in various pathological conditions.
TrkA is one of the high-affinity receptors for NGF typically expressed in neuronal cells. However, in cancer, aberrant TrkA expression has been observed in various types of tumors, including ovarian cancer, pancreatic cancer, and neuroblastoma [91,164]. In some cases, overexpression or activation of TrkA has been linked to tumor growth, survival, and angiogenesis [165]. Furthermore, higher levels of NGF and TrkA have often been associated with tumor perineural invasion (PNI) [110]. TrkB and TrkC also interact with neurotrophins like NGF and aberrant expression has been reported in different cancers [112]. TrkB is associated with aggressive tumor behavior, metastasis, and resistance to chemotherapy in some cases [24]. The p75NTR is a low-affinity receptor for NGF that, unlike Trk receptors, does not have intrinsic kinase activity. It can interact with various NTs, including NGF. The above-mentioned Trk signaling transduction and activation might also proceed in tumor cells when NTRK gene changes occur in these cells. On the whole, some types of gene variations associated with NTRK are known. The most common one in tumors is the NTRK gene fusion. Indeed, when the 3′ part of NTRK fuses with the 5′ part of another gene, the equivalent products of expression could elicit a subsequent oncogenic signal and ligand-independent kinase activation [234,235].
Trk fusion is widely distributed in several cancer types. The most evident cases are congenital fibrosarcoma and mammary analog secretory carcinoma, with 100% NTRK fusion discovered in these two cancers [236,237,238]. Several studies demonstrated that NTRK gene fusions are influential factors in some acute myeloid leukemias [239,240]. Furthermore, NTRK fusion has also been shown in other tumors, such as colorectal cancer and lung adenocarcinoma [241].
Regardless of gene fusion, the NTRK copy upregulation in nerve cells could also elicit an atypical overexpression of Trk kinases. Indeed, it has been shown that TrkA and phospho-TrkA in MDA-MB-231 cells were distinctly potentiated compared to those in standard breast tissues [152]. It was also demonstrated that TrkA overexpression was highly associated with tumor metastasis and cell proliferation [242]. Although Trk mutations in a single point are relatively rare during cancer development, several reports debated that Trk mutations in a single amino acid were associated with drug resistance versus Trk kinase inhibitors [235,243].
In cancer, p75NTR expression has been reported in multiple tumor types, including breast cancer, prostate cancer, and glioma with a complex and context-dependent role [9,26,37,244]. It can have both pro-survival and pro-apoptotic effects, depending on the cellular context and the presence of other co-receptors or ligands [85,136]. In some cases, p75NTR signaling has been associated with promoting cancer cell migration, invasion, and resistance to therapy.
Understanding the expression and function of NGF receptors, including both Trk receptors and p75NTR, is crucial for unraveling their roles in cancer development and progression. Researchers are actively investigating these receptors as potential therapeutic targets for cancer treatment, and some clinical trials are exploring the use of drugs targeting Trk receptors to inhibit tumor growth in specific cancer types with aberrant NGF receptor expression.

5. Epigenetics

Epigenetics refers to heritable changes in gene expression that occur without alterations in the DNA sequence itself, including methylation and histone modifications [245]. Epigenetic mechanisms play a significant role in regulating the inflammatory response, and conversely, inflammatory signals can induce changes in epigenetic marks [246]. Dysregulation of epigenetic mechanisms can contribute to chronic inflammation by altering the expression of genes involved in regulating the inflammatory response, while this chronic inflammation, in turn, is associated with aging and various diseases, including autoimmune disorders and cancer [247,248,249]. Furthermore, epigenetic modifications are critical in controlling the differentiation of immune cells so that changes in epigenetic marks can affect the function and response of immune cells, influencing the inflammatory state. On the other hand, epigenetic modifications play a pivotal role in tumor development and progression [10,250]. DNA methylation involves the addition of a methyl group to DNA, often occurring at specific sites called CpG islands [245]. Hypermethylation of promoter regions can silence tumor suppressor genes, while hypomethylation can activate oncogenes, both contributing to tumor formation. The chromatin, made of DNA and histones, can be modified through various mechanisms (e.g., acetylation, methylation, phosphorylation). ATP-dependent chromatin remodeling complexes alter the chromatin structure, impacting gene expression, while alterations in histone modifications can influence gene expression by changing how tightly DNA is packaged around histones, affecting the accessibility of genes to transcription machinery so that dysregulation of histone modifications can contribute to tumor initiation and progression [251]. MicroRNAs (miRNAs) and long non-coding RNAs (lncRNAs) are involved in post-transcriptional gene regulation and can influence various cellular processes, including proliferation, apoptosis, and differentiation, contributing to tumor development [252,253]. Epigenetic changes contribute to tumor heterogeneity, where different cells within a tumor have varying genetic and epigenetic profiles, affecting treatment responses. Studies suggest that NGF can regulate the expression of DNA methyltransferases (DNMTs), enzymes responsible for adding methyl groups to DNA [254,255]. Altered DNMT activity due to NGF signaling can impact gene expression patterns, influencing neuronal development and function. Actually, many inflammatory mediators like tumor necrosis factor alpha (TNF-α) and interleukin-6 (IL-6) can influence the expression of DNMTs and alter chromatin’s histone [256,257]. These findings have clinical significance; an example is nonsteroidal anti-inflammatory drugs (NSAIDs), which can participate in various signaling pathways [104]. A recent study demonstrated that ibuprofen epigenetically increases p75NTR expression by downregulating promoter methylation and upregulating m6A-RNA-methylation in SGC7901 and MKN45 cells [258].
NGF signaling also affects histone modifications, inducing changes in histone acetylation and methylation, altering chromatin structure and gene accessibility to modulate gene expression during neuronal differentiation and plasticity [259]. NGF signaling can regulate the expression of specific miRNAs, modulating the expression of genes involved in cell growth, differentiation, and survival. Dysregulation of NGF signaling and associated epigenetic changes have been implicated in neurological and tumoral disorders [51,120,201]. Drugs targeting epigenetic modifications (e.g., DNAMT inhibitors, histone deacetylase inhibitors) aim to reverse aberrant epigenetic changes and restore normal gene expression, showing promise in cancer treatment [260]. Understanding the interplay between NGF and epigenetics holds promise for developing novel therapeutic strategies targeting epigenetic mechanisms to modulate NGF signaling in various conditions. Epigenetic signatures associated with inflammation-related diseases could serve as biomarkers for diagnosis, prognosis, and the development of personalized treatments.

6. Therapeutic Potential of Neurotrophins and Their Receptors in Tumors

Many have suggested effective approaches to inhibit cancer via NGF inhibition, suggesting a promising therapeutic potential [160,161]. Studies on anti-NGF treatments in mouse models showed that NGF impacts tumor progression and metastasis in a temporally dependent manner [157].
Since the identification of tyrosine kinase receptors (TRKs) in the 1960s and their subsequent categorization into distinct families, their significant involvement in various facets of cellular biology has become increasingly evident [135]. Simultaneously, it has become clear that aberrations in TRKs can significantly influence a wide range of cellular processes, including tumor development, shaping their biological behavior from the early stages to advanced disease progression. The identification of mutations in TRKs has paved the way for the development of novel pharmaceuticals designed to inhibit these receptors and their downstream pathways, signifying a groundbreaking advancement in cancer therapy with the potential to impact mortality rates. Consequently, numerous inhibitors targeting TRK receptors are now routinely employed in the treatment of diverse human malignancies. Moreover, ongoing molecular research is actively exploring new mutations and drug candidates for cancer therapy, aiming to enhance patient quality of life and overall survival.
As stated before, NGF in the TME plays a multifaceted role, impacting various aspects of the immune response and playing a role in drug resistance in various types of cancers, contributing to the growth of inherently resistant populations and influencing the response of initially drug-sensitive cells. Enhanced NGF-TrkA signaling can promote cell proliferation through the PI3K/Akt and MAPK/ERK pathways, and sometimes, it has been associated with pro-survival signaling through p75NTR, preventing apoptosis triggered by chemotherapy drugs and allowing inherently resistant cells to survive and proliferate despite treatment [19,117,261]. Furthermore, NGF and pro-NGF may induce a state of quiescence in cancer cells, leading to cells not actively dividing, which are less susceptible to drugs that target rapidly dividing cells [232]. On the other hand, prolonged exposure to chemotherapy can lead to alterations in the TME, inducing increased NGF secretion by neighboring cells or stromal cells, which in turn could facilitate adaptive resistance mechanisms in sensitive cells (activation of survival pathways, DNA repair mechanisms, epigenetic changes), allowing cells to cope with the stress induced by chemotherapy and survive its effects [9,180]. Blocking NGF and its receptors, or the involved pathways, alongside chemotherapy could sensitize cancer cells to chemotherapy, preventing the development of drug resistance and possibly helping overcome or delay the emergence of drug resistance by targeting multiple pathways simultaneously [262,263]. Furthermore, inhibiting NGF or its receptors might enhance the efficacy of immunotherapies by reducing immunosuppression within the TME and preventing T cell dysfunction. The role of NGF and its receptor inhibitors currently seems limited in reducing symptomatology (mostly neuropathy) associated with chemotherapy [264].
The NTRK gene family encompasses three distinct members, namely NTRK1, NTRK2, and NTRK3, each responsible for producing TrkA, TrkB, and TrkC proteins. Rearrangement of these genes can occur either within the same chromosome or between different chromosomes, resulting in the fusion of two genes. This fusion event has the potential to create abnormal Trk proteins, ultimately promoting uncontrolled cellular proliferation [265]. The fusion of Trk genes gives rise to hybrid proteins that retain their functional tyrosine kinase domains, causing a sustained activation of downstream signaling pathways. This phenomenon is implicated in the development of various cancer types. Currently, there is ongoing research into inhibitors that exhibit promising tolerability and effectiveness, being evaluated in specific clinical trials across a range of cancer cell types.
To avoid NGF-related side effects, researchers have explored methods to stimulate the synthesis and release of endogenous NGF near damaged tissue, offering a potential avenue for therapeutic intervention.

7. Discussion

In this narrative review, we comprehensively explored the intricate molecular mechanisms through which inflammation and NGF impact tumor cell growth, survival, and death. We delved into the signaling pathways activated by inflammation and NGF, including the role in the microenvironment, interaction with the immune system, and the various molecules that play a role in tumorigenesis. NGF and inflammation have many meeting points and often act in a unanimous way to induce cancer. On the other hand, NGF is a complex molecule that has a pro-inflammatory and pro-tumoral role but also a negative feedback able to inhibit excessive inflammatory responses and tumor progression. The role of NGF is determined by the specific context and by the high and low expression of its specific receptors by the cells. By understanding the downstream effectors and cross-talk with other signaling cascades, we can gain deeper insights into the regulation of tumor cell proliferation and survival by NGF. Furthermore, we shed light on the multifaceted role of NGF in promoting tumor angiogenesis, its influence on EMT, and its ability to modulate the immune response within the TME. The reciprocal interactions between cancer cells and the surrounding stromal and immune cells mediated by NGF highlight its potential as a promising target for cancer therapeutics. However, the paradoxical effects of NGF in cancer extend beyond its oncogenic potential, as accumulating evidence suggests its involvement in promoting tumor cell death in specific cellular contexts. Understanding the factors that drive this pro-apoptotic activity will be crucial for the development of novel therapeutic approaches that harness NGF-induced cell death pathways to combat cancer progression. Indeed, NGF plays a multifaceted role in human health, ranging from its essential functions in the nervous system to its potential implications in promoting cell proliferation and survival in cancer cells. Further research is needed to fully understand the extent of NGF’s involvement in tumors and its potential therapeutic applications, especially in light of its role as a peculiar factor of the inflammatory process.

8. Conclusions

In conclusion, this report may provide a comprehensive overview of the multifaceted role of NGF as a component of the inflammatory phenomena in tumor cell growth and death, offering new perspectives on its potential as a therapeutic target in cancer treatment. The insights gained from unraveling the intricate interactions of NGF, inflammatory molecules, and the TME may pave the way for the development of precision therapies, enabling the exploitation of its dual nature for more effective and personalized cancer interventions.

Author Contributions

Conceptualization, S.T., G.F. and M.F.; methodology, S.T., G.F., P.T., M.L., F.F., B.C., L.C., A.P. and M.F.; validation, L.T., F.F., S.A. and A.G.; formal analysis, S.T., G.F. and M.F.; resources, S.T., G.F. and M.F.; data curation, L.T., F.F., S.A., B.C., P.T. and A.G.; writing—original draft preparation, S.T.; writing—review and editing, S.T., G.F. and M.F.; supervision, S.T., G.F., P.T., M.L., F.F., A.P., M.F., L.T., L.C., B.C. and A.G. 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 thank Sapienza Università di Roma, Italy, and IBBC-CNR of Rome, Italy.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Tian, J.; Han, Z.; Song, D.; Peng, Y.; Xiong, M.; Chen, Z.; Duan, S.; Zhang, L. Engineered Exosome for Drug Delivery: Recent Development and Clinical Applications. Int. J. Nanomed. 2023, 18, 7923–7940. [Google Scholar] [CrossRef] [PubMed]
  2. Mir, S.A.; Dar, A.; Hamid, L.; Nisar, N.; Malik, J.A.; Ali, T.; Nabi Bader, G. Flavonoids as promising molecules in the cancer therapy: An insight. Curr. Res. Pharmacol. Drug Discov. 2024, 6, 100167. [Google Scholar] [CrossRef] [PubMed]
  3. Thiery, J.; Fahrner, M. Integration of proteomics in the molecular tumor board. Proteomics 2023, e2300002. [Google Scholar] [CrossRef] [PubMed]
  4. Verma, C.; Pawar, V.A.; Srivastava, S.; Tyagi, A.; Kaushik, G.; Shukla, S.K.; Kumar, V. Cancer Vaccines in the Immunotherapy Era: Promise and Potential. Vaccines 2023, 11, 1783. [Google Scholar] [CrossRef] [PubMed]
  5. De Raffele, D.; Ilie, I.M. Unlocking novel therapies: Cyclic peptide design for amyloidogenic targets through synergies of experiments, simulations, and machine learning. Chem. Commun. 2023, 60, 632–645. [Google Scholar] [CrossRef]
  6. Chen, Y.; Wang, X.; Ye, Y.; Ren, Q. Gut microbiota in cancer: Insights on microbial metabolites and therapeutic strategies. Med. Oncol. 2023, 41, 25. [Google Scholar] [CrossRef]
  7. Liao, C.P.; Booker, R.C.; Brosseau, J.P.; Chen, Z.; Mo, J.; Tchegnon, E.; Wang, Y.; Wade Clapp, D.; Le, L.Q. Contributions of inflammation and tumor microenvironment to neurofibroma tumorigenesis. J. Clin. Investig. 2018, 128, 2848–2861. [Google Scholar] [CrossRef]
  8. Zhao, H.; Wu, L.; Yan, G.; Chen, Y.; Zhou, M.; Wu, Y.; Li, Y. Inflammation and tumor progression: Signaling pathways and targeted intervention. Signal Transduct. Target Ther. 2021, 6, 263. [Google Scholar] [CrossRef]
  9. Triaca, V.; Carito, V.; Fico, E.; Rosso, P.; Fiore, M.; Ralli, M.; Lambiase, A.; Greco, A.; Tirassa, P. Cancer stem cells-driven tumor growth and immune escape: The Janus face of neurotrophins. Aging 2019, 11, 11770–11792. [Google Scholar] [CrossRef]
  10. Ferraguti, G.; Terracina, S.; Petrella, C.; Greco, A.; Minni, A.; Lucarelli, M.; Agostinelli, E.; Ralli, M.; de Vincentiis, M.; Raponi, M.; et al. Alcohol and Head and Neck Cancer: Updates on the Role of Oxidative Stress, Genetic, Epigenetics, Oral Microbiota, Antioxidants, and Alkylating Agents. Antioxidants 2022, 11, 145. [Google Scholar] [CrossRef]
  11. Aygun, N. Biological and Genetic Features of Neuroblastoma and Their Clinical Importance. Curr. Pediatr. Rev. 2018, 14, 73–90. [Google Scholar] [CrossRef] [PubMed]
  12. Singh, R.; Karri, D.; Shen, H.; Shao, J.; Dasgupta, S.; Huang, S.; Edwards, D.E.; Ittmann, M.M.; O’Malley, B.W.; Yi, P. TRAF4-mediated ubiquitination of NGF receptor TrkA regulates prostate cancer metastasis. J. Clin. Investig. 2018, 128, 3129–3143. [Google Scholar] [CrossRef] [PubMed]
  13. Jiang, J.; Bai, J.; Qin, T.; Wang, Z.; Han, L. NGF from pancreatic stellate cells induces pancreatic cancer proliferation and invasion by PI3K/AKT/GSK signal pathway. J. Cell Mol. Med. 2020, 24, 5901–5910. [Google Scholar] [CrossRef] [PubMed]
  14. Minnone, G.; De Benedetti, F.; Bracci-Laudiero, L. NGF and its receptors in the regulation of inflammatory response. Int. J. Mol. Sci. 2017, 18, 1028. [Google Scholar] [CrossRef] [PubMed]
  15. Peng, T.; Guo, Y.; Gan, Z.; Ling, Y.; Xiong, J.; Liang, X.; Cui, J. Nerve Growth Factor (NGF) Encourages the Neuroinvasive Potential of Pancreatic Cancer Cells by Activating the Warburg Effect and Promoting Tumor Derived Exosomal miRNA-21 Expression. Oxid. Med. Cell Longev. 2022, 2022, 8445093. [Google Scholar] [CrossRef]
  16. Zuo, J.; Yi, C.; Chen, Z.; Zhou, B.; Yang, T.; Lin, J. A novel refined pyroptosis and inflammasome-related genes signature for predicting prognosis and immune microenvironment in pancreatic ductal adenocarcinoma. Sci. Rep. 2022, 12, 18384. [Google Scholar] [CrossRef]
  17. Anagnostopoulou, V.; Pediaditakis, I.; Alkahtani, S.; Alarifi, S.A.; Schmidt, E.M.; Lang, F.; Gravanis, A.; Charalampopoulos, I.; Stournaras, C. Differential effects of dehydroepiandrosterone and testosterone in prostate and colon cancer cell apoptosis: The role of nerve growth factor (NGF) receptors. Endocrinology 2013, 154, 2446–2456. [Google Scholar] [CrossRef]
  18. Bono, F.; Lamarche, I.; Bornia, J.; Savi, P.; Della Valle, G.; Herbert, J.M. Nerve growth factor (NGF) exerts its pro-apoptotic effect via the P75(NTR) receptor in a cell cycle-dependent manner. FEBS Lett. 1999, 457, 93–97. [Google Scholar] [CrossRef]
  19. Molloy, N.H.; Read, D.E.; Gorman, A.M. Nerve growth factor in cancer cell death and survival. Cancers 2011, 3, 510–530. [Google Scholar] [CrossRef]
  20. Fiore, M.; Chaldakov, G.N.; Aloe, L. Nerve growth factor as a signaling molecule for nerve cells and also for the neuroendocrine-immune systems. Rev. Neurosci. 2009, 20, 133–145. [Google Scholar] [CrossRef]
  21. Lykissas, M.G.; Batistatou, A.K.; Charalabopoulos, K.A.; Beris, A.E. The role of neurotrophins in axonal growth, guidance, and regeneration. Curr. Neurovasc. Res. 2007, 4, 143–151. [Google Scholar] [CrossRef] [PubMed]
  22. Rocco, M.L.; Soligo, M.; Manni, L.; Aloe, L. Nerve Growth Factor: Early Studies and Recent Clinical Trials. Curr. Neuropharmacol. 2018, 16, 1455–1465. [Google Scholar] [CrossRef] [PubMed]
  23. Shooter, E.M. Early days of the nerve growth factor proteins. Annu. Rev. Neurosci. 2001, 24, 601–629. [Google Scholar] [CrossRef]
  24. Schramm, A.; Schulte, J.H.; Astrahantseff, K.; Apostolov, O.; Van Limpt, V.; Sieverts, H.; Kuhfittig-Kulle, S.; Pfeiffer, P.; Versteeg, R.; Eggert, A. Biological effects of TrkA and TrkB receptor signaling in neuroblastoma. Cancer Lett. 2005, 228, 143–153. [Google Scholar] [CrossRef]
  25. Huang, E.J.; Reichardt, L.F. Trk Receptors: Roles in Neuronal Signal Transduction. Annu. Rev. Biochem. 2003, 72, 609–642. [Google Scholar] [CrossRef]
  26. Bradshaw, R.A.; Pundavela, J.; Biarc, J.; Chalkley, R.J.; Burlingame, A.L.; Hondermarck, H. NGF and ProNGF: Regulation of neuronal and neoplastic responses through receptor signaling. Adv. Biol. Regul. 2015, 58, 16–27. [Google Scholar] [CrossRef]
  27. Baldassarro, V.A.; Cescatti, M.; Rocco, M.L.; Aloe, L.; Lorenzini, L.; Giardino, L.; Calzà, L. Nerve growth factor promotes differentiation and protects the oligodendrocyte precursor cells from in vitro hypoxia/ischemia. Front. Neurosci. 2023, 17, 1111170. [Google Scholar] [CrossRef]
  28. Ferraguti, G.; Terracina, S.; Micangeli, G.; Lucarelli, M.; Tarani, L.; Ceccanti, M.; Spaziani, M.; D’Orazi, V.; Petrella, C.; Fiore, M. NGF and BDNF in pediatrics syndromes. Neurosci. Biobehav. Rev. 2023, 145, 105015. [Google Scholar] [CrossRef]
  29. Fahnestock, M.; Yu, G.; Coughlin, M.D. ProNGF: A neurotrophic or an apoptotic molecule? Prog. Brain Res. 2004, 146, 101–110. [Google Scholar] [CrossRef]
  30. Mohamed, R.; Coucha, M.; Elshaer, S.L.; Artham, S.; Lemtalsi, T.; El-Remessy, A.B. Inducible overexpression of endothelial proNGF as a mouse model to study microvascular dysfunction. Biochim. Biophys. Acta—Mol. Basis Dis. 2018, 1864, 746–757. [Google Scholar] [CrossRef]
  31. Song, L.; Xu, L.F.; Pu, Z.X.; Wang, H.H. HHIL-10 inhibits apoptosis in brain tissue around the hematoma after ICH by inhibiting proNGF. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 3005–3011. [Google Scholar] [CrossRef] [PubMed]
  32. Minnone, G.; Soligo, M.; Caiello, I.; Prencipe, G.; Manni, L.; Marafon, D.P.; Magni-Manzoni, S.; Manzo, A.; De Benedetti, F.; Gracci-Laudiero, L. ProNGF-p75NTR axis plays a proinflammatory role in inflamed joints: A novel pathogenic mechanism in chronic arthritis. RMD Open 2017, 3, e000441. [Google Scholar] [CrossRef] [PubMed]
  33. Barcelona, P.F.; Sitaras, N.; Galan, A.; Esquiva, G.; Jmaeff, S.; Jian, Y.; Sarunic, M.V.; Cuenca, N.; Sapieha, P.; Saragovi, H.U. p75NTR and its ligand ProNGF activate paracrine mechanisms etiological to the vascular, inflammatory, and neurodegenerative pathologies of diabetic retinopathy. J. Neurosci. 2016, 36, 8826–8841. [Google Scholar] [CrossRef]
  34. Capsoni, S.; Brandi, R.; Arisi, I.; D’Onofrio, M.; Cattaneo, A. A Dual Mechanism Linking NGF/proNGF Imbalance and Early Inflammation to Alzheimer’s Disease Neurodegeneration in the AD11 Anti-NGF Mouse Model. CNS Neurol. Disord—Drug Targets 2012, 10, 635–647. [Google Scholar] [CrossRef] [PubMed]
  35. Descamps, S.; Toillon, R.A.; Adriaenssens, E.; Pawlowski, V.; Cool, S.M.; Nurcombe, V.; Le Bourhis, X.; Boilly, B.; Peyrat, J.P.; Hondermarck, H. Nerve Growth Factor Stimulates Proliferation and Survival of Human Breast Cancer Cells through Two Distinct Signaling Pathways. J. Biol. Chem. 2001, 276, 17864–17870. [Google Scholar] [CrossRef] [PubMed]
  36. Meco, D.; Di Francesco, A.M.; Melotti, L.; Ruggiero, A.; Riccardi, R. Ectopic nerve growth factor prevents proliferation in glioma cells by senescence induction. J. Cell Physiol. 2019, 234, 6820–6830. [Google Scholar] [CrossRef] [PubMed]
  37. Chopin, V.; Lagadec, C.; Toillon, R.A.; Le Bourhis, X. Neurotrophin signaling in cancer stem cells. Cell Mol. Life Sci. 2016, 73, 1859–1870. [Google Scholar] [CrossRef]
  38. Marsland, M.; Dowdell, A.; Jiang, C.C.; Wilmott, J.S.; Scolyer, R.A.; Zhang, X.D.; Hondermarck, H.; Faulkner, S. Expression of NGF/proNGF and Their Receptors TrkA, p75NTR and Sortilin in Melanoma. Int. J. Mol. Sci. 2022, 23, 4260. [Google Scholar] [CrossRef]
  39. Peach, C.J.; Tonello, R.; Gomez, K.; Calderon-Rivera, A.; Sánchez, M.R.; Maile, L.; Perez-Miller, S.; Manu, A.M.; Hahn, H.; Thomsen, A.R.B.; et al. Neuropilin-1 is a co-receptor for NGF and TrkA-evoked pain. BioRxiv 2023. [Google Scholar] [CrossRef]
  40. Pond, A.; Roche, F.K.; Letourneau, P.C. Temporal regulation of neuropilin-1 expression and sensitivity to semaphorin 3A in NGF- and NT3-responsive chick sensory neurons. J. Neurobiol. 2002, 51, 43–53. [Google Scholar] [CrossRef]
  41. Reza, J.N.; Gavazzi, I.; Cohen, J. Neuropilin-1 is expressed on adult mammalian dorsal root ganglion neurons and mediates semaphorin3a/collapsin-1-induced growth cone collapse by small diameter sensory afferents. Mol. Cell Neurosci. 1999, 14, 317–326. [Google Scholar] [CrossRef] [PubMed]
  42. Claudio Cuello, A.; Pentz, R.; Hall, H. The brain NGF metabolic pathway in health and in Alzheimer’s pathology. Front. Neurosci. 2019, 13, 62. [Google Scholar] [CrossRef] [PubMed]
  43. Hristova, M.; Aloe, L. Metabolic syndrome—Neurotrophic hypothesis. Med. Hypotheses 2006, 66, 545–549. [Google Scholar] [CrossRef] [PubMed]
  44. Alkhadar, H.; Macluskey, M.; White, S.; Ellis, I. Nerve growth factor-induced migration in oral and salivary gland tumour cells utilises the PI3K/Akt signalling pathway: Is there a link to perineural invasion? J. Oral Pathol. Med. 2020, 49, 227–234. [Google Scholar] [CrossRef]
  45. Smith, M.A. Hippocampal vulnerability to stress and aging: Possible role of neurotrophic factors. Behav. Brain Res. 1996, 78, 25–36. [Google Scholar] [CrossRef] [PubMed]
  46. Miller, R.; King, M.A.; Heaton, M.B.; Walker, D.W. The effects of chronic ethanol consumption on neurotrophins and their receptors in the rat hippocampus and basal forebrain. Brain Res. 2002, 950, 137–147. [Google Scholar] [CrossRef] [PubMed]
  47. Lin, K.; Huang, J.; Luo, H.; Luo, C.; Zhu, X.; Bu, F.; Xiao, H.; Xiao, L.; Zhu, Z. Development of a prognostic index and screening of potential biomarkers based on immunogenomic landscape analysis of colorectal cancer. Aging 2020, 12, 5832–5857. [Google Scholar] [CrossRef] [PubMed]
  48. Ceci, F.M.; Ferraguti, G.; Petrella, C.; Greco, A.; Tirassa, P.; Iannitelli, A.; Ralli, M.; Vitali, M.; Ceccanti, M.; Chaldakov, G.N.; et al. Nerve Growth Factor, Stress and Diseases. Curr. Med. Chem. 2020, 28, 2943–2959. [Google Scholar] [CrossRef]
  49. D’Angelo, A.; Ceccanti, M.; Petrella, C.; Greco, A.; Tirassa, P.; Rosso, P.; Ralli, M.; Ferraguti, G.; Fiore, M.; Messina, M.P. Role of neurotrophins in pregnancy, delivery and postpartum. Eur. J. Obstet. Gynecol. Reprod. Biol. 2020, 247, 32–41. [Google Scholar] [CrossRef]
  50. Fiore, M.; Korf, J.; Antonelli, A.; Talamini, L.; Aloe, L. Long-lasting effects of prenatal MAM treatment on water maze performance in rats: Associations with altered brain development and neurotrophin levels. Neurotoxicol. Teratol. 2002, 24, 179–191. [Google Scholar] [CrossRef]
  51. Ciafrè, S.; Ferraguti, G.; Tirassa, P.; Iannitelli, A.; Ralli, M.; Greco, A.; Chaldakov, G.N.; Rosso, P.; Fico, E.; Messina, M.P.; et al. Nerve growth factor in the psychiatric brain. Riv. Psichiatr. 2020, 55, 4–15. [Google Scholar] [CrossRef] [PubMed]
  52. Terracina, S.; Ferraguti, G.; Tarani, L.; Fanfarillo, F.; Tirassa, P.; Ralli, M.; Iannella, G.; Polimeni, A.; Lucarelli, M.; Greco, A.; et al. Nerve Growth Factor and Autoimmune Diseases. Curr. Issues Mol. Biol. 2023, 45, 8950–8973. [Google Scholar] [CrossRef] [PubMed]
  53. Lebedev, T.; Vagapova, E.; Spirin, P.; Rubtsov, P.; Astashkova, O.; Mikheeva, A.; Sorokin, M.; Vladimirova, U.; Suntsova, M.; Konovalov, D.; et al. Growth factor signaling predicts therapy resistance mechanisms and defines neuroblastoma subtypes. Oncogene 2021, 40, 6258–6272. [Google Scholar] [CrossRef] [PubMed]
  54. Faulkner, S.; Griffin, N.; Rowe, C.W.; Jobling, P.; Lombard, J.M.; Oliveira, S.M.; Walker, M.M.; Hondermarck, H. Nerve growth factor and its receptor tyrosine kinase TrkA are overexpressed in cervical squamous cell carcinoma. FASEB BioAdv. 2020, 2, 398–408. [Google Scholar] [CrossRef] [PubMed]
  55. Aloe, L.; Simone, M.D.; Properzi, F. Nerve growth factor: A neurotrophin with activity on cells of the immune system. Microsc. Res. Tech. 1999, 45, 285–291. [Google Scholar] [CrossRef]
  56. Roe, K. An inflammation classification system using cytokine parameters. Scand. J. Immunol. 2021, 93, e12970. [Google Scholar] [CrossRef] [PubMed]
  57. Oronsky, B.; Caroen, S.; Reid, T. What Exactly Is Inflammation (and What Is It Not?). Int. J. Mol. Sci. 2022, 23, 14905. [Google Scholar] [CrossRef]
  58. Schmid-Schönbein, G.W. Analysis of inflammation. Annu. Rev. Biomed. Eng. 2006, 8, 93–151. [Google Scholar] [CrossRef]
  59. Varela, M.L.; Mogildea, M.; Moreno, I.; Lopes, A. Acute Inflammation and Metabolism. Inflammation 2018, 41, 1115–1127. [Google Scholar] [CrossRef]
  60. Deng, L.; He, S.; Guo, N.; Tian, W.; Zhang, W.; Luo, L. Molecular mechanisms of ferroptosis and relevance to inflammation. Inflamm. Res. 2023, 72, 281–299. [Google Scholar] [CrossRef]
  61. Tracy, R.P. The five cardinal signs of inflammation: Calor, dolor, rubor, tumor... and penuria (apologies to Aulus Cornelius Celsus, de medicina, c. A.D. 25). J. Gerontol. Ser. A Biol. Sci. Med. Sci. 2006, 61, 1051–1052. [Google Scholar] [CrossRef] [PubMed]
  62. Dandri, M.; Bertoletti, A.; Lütgehetmann, M. Innate immunity in hepatitis B and D virus infection: Consequences for viral persistence, inflammation, and T cell recognition. Semin. Immunopathol. 2021, 43, 535–548. [Google Scholar] [CrossRef] [PubMed]
  63. Cavaillon, J.M. Once upon a time, inflammation. J. Venom. Anim. Toxins Incl. Trop. Dis. 2021, 27, e20200147. [Google Scholar] [CrossRef] [PubMed]
  64. Arulselvan, P.; Fard, M.T.; Tan, W.S.; Gothai, S.; Fakurazi, S.; Norhaizan, M.E.; Kumar, S.S. Role of Antioxidants and Natural Products in Inflammation. Oxid. Med. Cell Longev. 2016, 2016, 1–15. [Google Scholar] [CrossRef] [PubMed]
  65. Ceci, F.M.; Ferraguti, G.; Petrella, C.; Greco, A.; Ralli, M.; Iannitelli, A.; Carito, V.; Tirassa, P.; Chaldakov, G.N.; Messina, M.P.; et al. Nerve Growth Factor in Alcohol Use Disorders. Curr. Neuropharmacol. 2020, 19, 45–60. [Google Scholar] [CrossRef] [PubMed]
  66. Muratori, L.; Lohse, A.W.; Lenzi, M. Diagnosis and management of autoimmune hepatitis. BMJ 2023, 380, e070201. [Google Scholar] [CrossRef] [PubMed]
  67. Sun, B.; Karin, M. Inflammation and liver tumorigenesis. Front. Med. 2013, 7, 242–254. [Google Scholar] [CrossRef] [PubMed]
  68. Ciafrè, S.; Ferraguti, G.; Greco, A.; Polimeni, A.; Ralli, M.; Ceci, F.M.; Ceccanti, M.; Fiore, M. Alcohol as an early life stressor: Epigenetics, metabolic, neuroendocrine and neurobehavioral implications. Neurosci. Biobehav. Rev. 2020, 118, 654–668. [Google Scholar] [CrossRef]
  69. Singh, N.; Baby, D.; Rajguru, J.; Patil, P.; Thakkannavar, S.; Pujari, V. Inflammation and cancer. Ann. Afr. Med. 2019, 18, 121–126. [Google Scholar] [CrossRef]
  70. Hernaez, R.; Solà, E.; Moreau, R.; Ginès, P. Acute-on-chronic liver failure: An update. Gut 2017, 66, 541–553. [Google Scholar] [CrossRef]
  71. Hunter, L.J.; Wood, D.M.; Dargan, P.I. The patterns of toxicity and management of acute nonsteroidal anti-inflammatory drug (NSAID) overdose. Open Access Emerg. Med. 2011, 3, 39–48. [Google Scholar] [CrossRef] [PubMed]
  72. Curtis, J.R.; Olivieri, J.; Allison, J.J.; Gaffo, A.; Juarez, L.; Kovac, S.H.; Person, S.; Saag, K.G. A group randomized trial to improve safe use of nonsteroidal anti-inflammatory drugs. Am. J. Manag. Care 2005, 11, 537–543. [Google Scholar] [PubMed]
  73. Slaats, J.; ten Oever, J.; van de Veerdonk, F.L.; Netea, M.G. IL-1β/IL-6/CRP and IL-18/ferritin: Distinct Inflammatory Programs in Infections. PLoS Pathog. 2016, 12, e1005973. [Google Scholar] [CrossRef] [PubMed]
  74. Aloe, L.; Rocco, M.L.; Balzamino, B.O.; Micera, A. Nerve growth factor: Role in growth, differentiation and controlling cancer cell development. J. Exp. Clin. Cancer Res. 2016, 35, 1–7. [Google Scholar] [CrossRef] [PubMed]
  75. Cohen, S.; Levi-Montalcini, R.; Hamburger, V. A Nerve Growth-Stimulating Factor Isolated from Sarcom As 37 and 180. Proc. Natl. Acad. Sci. USA 1954, 40, 1014–1018. [Google Scholar] [CrossRef] [PubMed]
  76. Zeliadt, N. Rita Levi-Montalcini: NGF, the prototypical growth factor. Proc. Natl. Acad. Sci. USA 2013, 110, 4873–4876. [Google Scholar] [CrossRef] [PubMed]
  77. Goda, M.; Atagi, S.; Amitani, K.; Hobara, N.; Kitamura, Y.; Kawasaki, H. Nerve growth factor suppresses prostate tumor growth. J. Pharmacol. Sci. 2010, 112, 463–466. [Google Scholar] [CrossRef] [PubMed]
  78. Krygier, S.; Djakiew, D. Neurotrophin receptor p75NTR suppresses growth and nerve growth factor-mediated metastasis of human prostate cancer cells. Int. J. Cancer 2002, 98, 1–7. [Google Scholar] [CrossRef]
  79. Srinivasan, R.; Zabuawala, T.; Huang, H.; Zhang, J.; Gulati, P.; Fernandez, S.; Colleen Karlo, J.; Landreth, G.E.; Leone, G.; Ostrowski, M.C. Erk1 and erk2 regulate endothelial cell proliferation and migration during mouse embryonic angiogenesis. PLoS ONE 2009, 4, e8283. [Google Scholar] [CrossRef]
  80. Lin, H.; Huang, H.; Yu, Y.; Chen, W.; Zhang, S.; Zhang, Y. Nerve growth factor regulates liver cancer cell polarity and motility. Mol. Med. Rep. 2021, 23, 288. [Google Scholar] [CrossRef]
  81. Tarani, L.; Ceci, F.M.; Carito, V.; Ferraguti, G.; Petrella, C.; Greco, A.; Ralli, M.; Minni, A.; Spaziani, M.; Isidori, A.M.; et al. Neuroimmune Dysregulation in Prepubertal and Adolescent Individuals Affected by Klinefelter Syndrome. Endocr. Metab Immune Disord. Drug Targets 2022, 22, 105–114. [Google Scholar] [CrossRef]
  82. Fiore, M.; Petrella, C.; Coriale, G.; Rosso, P.; Fico, E.; Ralli, M.; Greco, A.; de Vincentiis, M.; Minni, A.; Polimeni, A.; et al. Markers of Neuroinflammation in the Serum of Prepubertal Children with Fetal Alcohol Spectrum Disorders. CNS Neurol. Disord. Drug Targets 2022, 21, 854–868. [Google Scholar] [CrossRef] [PubMed]
  83. Tarani, L.; Carito, V.; Ferraguti, G.; Petrella, C.; Greco, A.; Ralli, M.; Messina, M.P.; Rasio, D.; De Luca, E.; Putotto, C.; et al. Neuroinflammatory Markers in the Serum of Prepubertal Children with down Syndrome. J. Immunol. Res. 2020, 2020, 6937154. [Google Scholar] [CrossRef]
  84. Kritas, S.K.; Saggini, A.; Cerulli, G.; Caraffa, A.; Antinolfi, P.; Pantalone, A.; Frydas, S.; Rosati, M.; Tei, M.; Speziali, A.; et al. Neuropeptide NGF mediates neuro-immune response and inflammation through mast cell activation. J. Biol. Regul. Homeost Agents 2014, 28, 177–181. [Google Scholar] [PubMed]
  85. Bracci-Laudiero, L.; De Stefano, M.E. NGF in early embryogenesis, differentiation, and pathology in the nervous and immune systems. Curr. Top. Behav. Neurosci. 2016, 29, 125–152. [Google Scholar] [CrossRef]
  86. Vega, J.A.; García-Suárez, O.; Hannestad, J.; Pérez-Pérez, M.; Germanà, A. Neurotrophins and the immune system. J. Anat. 2003, 203, 1–19. [Google Scholar] [CrossRef]
  87. Seidel, M.F.; Hügle, T.; Morlion, B.; Koltzenburg, M.; Chapman, V.; Maassen Van Den Brink, A.; Lane, N.E.; Perrot, S.; Zieglgänsberger, W. Neurogenic inflammation as a novel treatment target for chronic pain syndromes. Exp. Neurol. 2022, 356, 114108. [Google Scholar] [CrossRef]
  88. Schumacher, M.A. Peripheral Neuroinflammation and Pain: How Acute Pain Becomes Chronic. Curr. Neuropharmacol. 2023, 22, 6–14. [Google Scholar] [CrossRef]
  89. Nayak, D.; Roth, T.L.; McGavern, D.B. Microglia development and function. Annu. Rev. Immunol. 2014, 32, 367–402. [Google Scholar] [CrossRef]
  90. Rizzi, C.; Tiberi, A.; Giustizieri, M.; Marrone, M.C.; Gobbo, F.; Carucci, N.M.; Meli, G.; Arisi, I.; D’Onofrio, M.; Marinelli, S.; et al. NGF steers microglia toward a neuroprotective phenotype. Glia 2018, 66, 1395–1416. [Google Scholar] [CrossRef]
  91. Micangeli, G.; Menghi, M.; Profeta, G.; Tarani, F.; Mariani, A.; Petrella, C.; Barbato, C.; Ferraguti, G.; Ceccanti, M.; Tarani, L.; et al. The Impact of Oxidative Stress on Pediatrics Syndromes. Antioxidants 2022, 11, 1983. [Google Scholar] [CrossRef] [PubMed]
  92. Saloman, J.L.; Singhi, A.D.; Hartman, D.J.; Normolle, D.P.; Albers, K.M.; Davis, B.M. Systemic Depletion of Nerve Growth Factor Inhibits Disease Progression in a Genetically Engineered Model of Pancreatic Ductal Adenocarcinoma. Pancreas 2018, 47, 856–863. [Google Scholar] [CrossRef] [PubMed]
  93. Carito, V.; Ceccanti, M.; Tarani, L.; Ferraguti, G.; Chaldakov, N.G.; Fiore, M. Neurotrophins’ Modulation by Olive Polyphenols. Curr. Med. Chem. 2016, 23, 3189–3197. [Google Scholar] [CrossRef] [PubMed]
  94. Sofroniew, M.V.; Howe, C.L.; Mobley, W.C. Nerve Growth Factor Signaling, Neuroprotection, and Neural Repair. Annu. Rev. Neurosci. 2001, 24, 1217–1281. [Google Scholar] [CrossRef]
  95. Tafreshi, A.P. Nerve growth factor prevents demyelination, cell death and progression of the disease in experimental allergic encephalomyelitis. Iran. J. Allergy Asthma Immunol. 2006, 5, 177–181. [Google Scholar]
  96. Allen, S.J.; Watson, J.J.; Shoemark, D.K.; Barua, N.U.; Patel, N.K. GDNF, NGF and BDNF as therapeutic options for neurodegeneration. Pharmacol. Ther. 2013, 138, 155–175. [Google Scholar] [CrossRef]
  97. Castrén, E.; Tanila, H. Neurotrophins and Dementia-Keeping in Touch. Neuron 2006, 51, 1–3. [Google Scholar] [CrossRef]
  98. Iulita, M.F.; Cuello, A.C. Nerve growth factor metabolic dysfunction in Alzheimer’s disease and Down syndrome. Trends Pharmacol. Sci. 2014, 35, 338–348. [Google Scholar] [CrossRef]
  99. Chao, M.Y.; Rajagopal, R.; Lee, F.S. Neurotrophin signalling in health and disease. Clin. Sci. 2006, 110, 167–173. [Google Scholar] [CrossRef]
  100. Mierke, C.T. The fundamental role of mechanical properties in the progression of cancer disease and inflammation. Rep. Prog. Phys. 2014, 77, 076602. [Google Scholar] [CrossRef]
  101. Allavena, P.; Garlanda, C.; Borrello, M.G.; Sica, A.; Mantovani, A. Pathways connecting inflammation and cancer. Curr. Opin. Genet. Dev. 2008, 18, 3–10. [Google Scholar] [CrossRef] [PubMed]
  102. Hagerling, C.; Casbon, A.J.; Werb, Z. Balancing the innate immune system in tumor development. Trends Cell Biol. 2015, 25, 214–220. [Google Scholar] [CrossRef]
  103. Reuter, S.; Gupta, S.C.; Chaturvedi, M.M.; Aggarwal, B.B. Oxidative stress, inflammation, and cancer: How are they linked? Free Radic. Biol. Med. 2010, 49, 1603–1616. [Google Scholar] [CrossRef] [PubMed]
  104. Todoric, J.; Antonucci, L.; Karin, M. Targeting inflammation in cancer prevention and therapy. Cancer Prev. Res. 2016, 9, 895–905. [Google Scholar] [CrossRef] [PubMed]
  105. Anderson, N.M.; Simon, M.C. The tumor microenvironment. Curr. Biol. 2020, 30, R921–R925. [Google Scholar] [CrossRef]
  106. Li, X.; Zhao, S.; Bian, X.; Zhang, L.; Lu, L.; Pei, S.; Dong, L.; Shi, W.; Huang, L.; Zhang, X.; et al. Signatures of EMT, immunosuppression, and inflammation in primary and recurrent human cutaneous squamous cell carcinoma at single-cell resolution. Theranostics 2022, 12, 7532–7549. [Google Scholar] [CrossRef]
  107. Greten, F.R.; Grivennikov, S.I. Inflammation and Cancer: Triggers, Mechanisms, and Consequences. Immunity 2019, 51, 27–41. [Google Scholar] [CrossRef]
  108. Quail, D.F.; Joyce, J.A. Microenvironmental regulation of tumor progression and metastasis. Nat. Med. 2013, 19, 1423–1437. [Google Scholar] [CrossRef]
  109. Liu, Y.; Cao, X. Characteristics and Significance of the Pre-metastatic Niche. Cancer Cell 2016, 30, 668–681. [Google Scholar] [CrossRef]
  110. Kuol, N.; Stojanovska, L.; Apostolopoulos, V.; Nurgali, K. Role of the Nervous System in Tumor Angiogenesis. Cancer Microenviron. 2018, 11, 1–11. [Google Scholar] [CrossRef]
  111. Levi-Montalcini, R.; Skaper, S.D.; Dal Toso, R.; Petrelli, L.; Leon, A. Nerve growth factor: From neurotrophin to neurokine. Trends Neurosci. 1996, 19, 514–520. [Google Scholar] [CrossRef] [PubMed]
  112. Romon, R.; Adriaenssens, E.; Lagadec, C.; Germain, E.; Hondermarck, H.; Le Bourhis, X. Nerve growth factor promotes breast cancer angiogenesis by activating multiple pathways. Mol. Cancer 2010, 9, 157. [Google Scholar] [CrossRef] [PubMed]
  113. Lazarovici, P.; Marcinkiewicz, C.; Lelkes, P.I. Cross talk between the cardiovascular and nervous systems: Neurotrophic effects of vascular endothelial growth factor (VEGF) and angiogenic effects of nerve growth factor (NGF)-implications in drug development. Curr. Pharm. Des. 2006, 12, 2609–2622. [Google Scholar] [CrossRef]
  114. Vera, C.; Tapia, V.; Vega, M.; Romero, C. Role of nerve growth factor and its TRKA receptor in normal ovarian and epithelial ovarian cancer angiogenesis. J. Ovarian Res. 2014, 7, 82. [Google Scholar] [CrossRef]
  115. Lambert, C.; Mathy-Hartert, M.; Dubuc, J.E.; Montell, E.; Vergés, J.; Munaut, C.; Noel, A.; Henrotin, Y. Characterization of synovial angiogenesis in osteoarthritis patients and its modulation by chondroitin sulfate. Arthritis Res. Ther. 2012, 14, R58. [Google Scholar] [CrossRef]
  116. Zhang, L.; Zhang, W.; Zhang, X.; Yihe, M.I.N.; Zhao, Y.; Wang, B.; Wei, L.; Mao, S.; Min, W. High-glucose microenvironment promotes perineural invasion of pancreatic cancer via activation of hypoxia inducible factor 1α. Oncol. Rep. 2022, 47, 64. [Google Scholar] [CrossRef]
  117. Han, S.; Wang, D.; Huang, Y.; Zeng, Z.; Xu, P.; Xiong, H.; Ke, Z.; Zhang, Y.; Hu, Y.; Wang, F.; et al. A reciprocal feedback between colon cancer cells and Schwann cells promotes the proliferation and metastasis of colon cancer. J. Exp. Clin. Cancer Res. 2022, 41, 348. [Google Scholar] [CrossRef]
  118. Whiteside, T.L. The tumor microenvironment and its role in promoting tumor growth. Oncogene 2008, 27, 5904–5912. [Google Scholar] [CrossRef]
  119. Jung, H.H.; Kim, J.Y.; Cho, E.Y.; Oh, J.M.; Lee, J.E.; Kim, S.W.; Nam, S.J.; Park, Y.H.; Ahn, J.S.; Im, Y.H.; et al. Elevated level of nerve growth factor (Ngf) in serum-derived exosomes predicts poor survival in patients with breast cancer undergoing neoadjuvant chemotherapy. Cancers 2021, 13, 5260. [Google Scholar] [CrossRef]
  120. Wang, W.; Li, L.; Chen, N.; Niu, C.; Li, Z.; Hu, J.; Cui, J. Nerves in the Tumor Microenvironment: Origin and Effects. Front. Cell Dev. Biol. 2020, 8, 601738. [Google Scholar] [CrossRef]
  121. Sun, L.; Chen, S.; Chen, M. Schwann Cells in the Tumor Microenvironment: Need More Attention. J. Oncol. 2022, 2022, 1–10. [Google Scholar] [CrossRef] [PubMed]
  122. Tetri, L.H.; Kolla, V.; Golden, R.L.; Iyer, R.; Croucher, J.L.; Choi, J.H.; Macfarland, S.P.; Naparaju, K.; Guan, P.; Nguyen, F.; et al. RET receptor expression and interaction with TRK receptors in neuroblastomas. Oncol. Rep. 2020, 44, 263–272. [Google Scholar] [CrossRef] [PubMed]
  123. Park, S.Y.; Yang, H.J.; Ye, M.; Liu, X.; Shim, I.; Chang, Y.T.; Bae, H. Neuroprotective effects of ex vivo-expanded regulatory T cells on trimethyltin-induced neurodegeneration in mice. J. Neuroinflamm. 2022, 19, 143. [Google Scholar] [CrossRef] [PubMed]
  124. Han, Y.; Liu, D.; Li, L. PD-1/PD-L1 pathway: Current researches in cancer. Am. J. Cancer Res. 2020, 10, 727–742. [Google Scholar] [PubMed]
  125. Shan, Q.; Takabatake, K.; Kawai, H.; Oo, M.W.; Sukegawa, S.; Fujii, M.; Nakano, K.; Nagatsuka, H. Crosstalk between cancer and different cancer stroma subtypes promotes the infiltration of tumor-associated macrophages into the tumor microenvironment of oral squamous cell carcinoma. Int. J. Oncol. 2022, 60, 78. [Google Scholar] [CrossRef] [PubMed]
  126. Zhang, Y.; Lin, C.; Liu, Z.; Sun, Y.; Chen, M.; Guo, Y.; Liu, W.; Zhang, C.; Chen, W.; Sun, J.; et al. Cancer cells co-opt nociceptive nerves to thrive in nutrient-poor environments and upon nutrient-starvation therapies. Cell Metab. 2022, 34, 1999–2017.e10. [Google Scholar] [CrossRef] [PubMed]
  127. Urzua, U.; Tapia, V.; Geraldo, M.P.; Selman, A.; Vega, M.; Romero, C. Nerve growth factor stimulates cellular proliferation of human epithelial ovarian cancer. Horm. Metab. Res. 2012, 44, 656–661. [Google Scholar] [CrossRef]
  128. Yang, L.; Shi, P.; Zhao, G.; Xu, J.; Peng, W.; Zhang, J.; Wang, X.; Dong, Z.; Chen, F.; Cui, H. Targeting cancer stem cell pathways for cancer therapy. Signal Transduct. Target Ther. 2020, 5, 8. [Google Scholar] [CrossRef]
  129. Zhang, P.; Sun, Y.; Ma, L. ZEB1: At the crossroads of epithelial-mesenchymal transition, metastasis and therapy resistance. Cell Cycle 2015, 14, 481–487. [Google Scholar] [CrossRef]
  130. Wu, Y.; Zhou, B.P. TNF-α/NFκ-B/Snail pathway in cancer cell migration and invasion. Br. J. Cancer 2010, 102, 639–644. [Google Scholar] [CrossRef]
  131. Scalia, P.; Williams, S.J.; Fujita-Yamaguchi, Y.; Giordano, A. Cell cycle control by the insulin-like growth factor signal: At the crossroad between cell growth and mitotic regulation. Cell Cycle 2023, 22, 1–37. [Google Scholar] [CrossRef] [PubMed]
  132. Tomellini, E.; Touil, Y.; Lagadec, C.; Julien, S.; Ostyn, P.; Ziental-Gelus, N.; Meigna, S.; Lengrand, J.; Adrianseen, E.; Palokowska, R.; et al. Nerve growth factor and prongf simultaneously promote symmetric self-renewal, quiescence, and epithelial to mesenchymal transition to enlarge the breast cancer stem cell compartment. Stem Cells 2015, 33, 342–353. [Google Scholar] [CrossRef] [PubMed]
  133. Di Donato, M.; Cernera, G.; Migliaccio, A.; Castoria, G. Nerve growth factor induces proliferation and aggressiveness in prostate cancer cells. Cancers 2019, 11, 784. [Google Scholar] [CrossRef] [PubMed]
  134. Marsland, M.; Dowdell, A.; Faulkner, S.; Jobling, P.; Rush, R.A.; Gedye, C.; Lynam, J.; Griffin, C.P.; Baker, M.; Marsland, J.; et al. ProNGF Expression and Targeting in Glioblastoma Multiforme. Int. J. Mol. Sci. 2023, 24, 1616. [Google Scholar] [CrossRef]
  135. Jiang, T.; Wang, G.; Liu, Y.; Feng, L.; Wang, M.; Liu, J.; Chen, Y.; Ouyang, L. Development of small-molecule tropomyosin receptor kinase (TRK) inhibitors for NTRK fusion cancers. Acta Pharm. Sin. B 2021, 11, 355–372. [Google Scholar] [CrossRef] [PubMed]
  136. Oelmann, E.; Sreter, L.; Schuller, I.; Serve, H.; Koenigsmann, M.; Wiedenmann, B.; Oderberg, D.; Reufi, B.; Thiel, E.; Berdel, W.E. Nerve Growth Factor Stimulates Clonal Growth of Human Lung Cancer Cell Lines and a Human Glioblastoma Cell Line Expressing High-Affinity Nerve Growth Factor Binding Sites Involving Tyrosine Kinase Signaling. Cancer Res. 1995, 55, 2212–2219. [Google Scholar] [PubMed]
  137. Singer, H.S.; Hansen, B.; Martinie, D.; Karp, C.L. Mitogenesis in glioblastoma multiforme cell lines: A role for NGF and its TrkA receptors. J. Neurooncol. 1999, 45, 1–8. [Google Scholar] [CrossRef]
  138. Forsyth, P.A.; Krishna, N.; Lawn, S.; Valadez, J.G.; Qu, X.; Fenstermacher, D.A.; Fournier, M.; Potthast, L.; Chinnaiyan, P.; Gibney, G.T.; et al. P75 Neurotrophin Receptor Cleavage By A- and Γ-Secretases Is Required for Neurotrophin-Mediated Proliferation of Brain Tumor-Initiating Cells. J. Biol. Chem. 2014, 289, 8067–8085. [Google Scholar] [CrossRef]
  139. Zhang, Z.; Yang, Y.; Gong, A.; Wang, C.; Liang, Y.; Chen, Y. Localization of NGF and TrkA at mitotic apparatus in human glioma cell line U251. Biochem. Biophys. Res. Commun. 2005, 337, 68–74. [Google Scholar] [CrossRef]
  140. Antonelli, A.; Chiaretti, A.; Piastra, M.; Vigneti, E.; Aloe, L. In vitro human ependymoblastoma cells differentiate after exposure to nerve growth factor. J. Neurocytol. 2004, 33, 503–515. [Google Scholar] [CrossRef]
  141. Engebraaten, O.; Bjerkvig, R.; Pedersen, P.-H.; Laerum, O.D. Effects of EGF, BFGF, NGF and PDGF(bb) on cell proliferative, migratory and invasive capacities of human brain-tumour biopsies In Vitro. Int. J. Cancer 1993, 53, 209–214. [Google Scholar] [CrossRef] [PubMed]
  142. Fanfarillo, F.; Ferraguti, G.; Lucarelli, M.; Francati, S.; Barbato, C.; Minni, A.; Ceccanti, M.; Tarani, L.; Petrella, C.; Fiore, M. The Impact of ROS and NGF in the Gliomagenesis and their Emerging Implications in the Glioma Treatment. CNS Neurol. Disord. Drug Targets 2023, 23, 449–462. [Google Scholar] [CrossRef] [PubMed]
  143. Chang, A.; Botteri, E.; Gillis, R.D.; Löfling, L.; Le, C.P.; Ziegler, A.I.; Chung, N.C.; Rowe, M.C.; Fabb, S.A.; Hartley, B.J.; et al. Beta-blockade enhances anthracycline control of metastasis in triple-negative breast cancer. Sci. Transl. Med. 2023, 15, eadf1147. [Google Scholar] [CrossRef] [PubMed]
  144. Descamp, S.; Pawlowski, V.; Révillion, F.; Hornez, L.; Hebbar, M.; Boilly, B.; Hondermarck, H.; Peyrat, J.P. Expression of nerve growth factor receptors and their prognostic value in human breast cancer. Cancer Res. 2001, 61, 4337–4340. [Google Scholar]
  145. Jin, M.; Wang, Y.; Zhou, T.; Li, W.; Wen, Q. Norepinephrine/β2-Adrenergic Receptor Pathway Promotes the Cell Proliferation and Nerve Growth Factor Production in Triple-Negative Breast Cancer. J. Breast Cancer 2023, 26, 268–285. [Google Scholar] [CrossRef] [PubMed]
  146. Tian, Y.; Qiu, X.; Qi, X.; Dong, Z.; Zhao, J.; Huang, J.; Xiang, J. Electroacupuncture promotes apoptosis and inhibits axonogenesis by activating p75 neurotrophin receptor for triple-negative breast xenograft in mice. J. Chem. Neuroanat. 2022, 124, 102133. [Google Scholar] [CrossRef] [PubMed]
  147. Bruno, F.; Arcuri, D.; Vozzo, F.; Malvaso, A.; Montesanto, A.; Maletta, R. Expression and Signaling Pathways of Nerve Growth Factor (NGF) and Pro-NGF in Breast Cancer: A Systematic Review. Curr. Oncol. 2022, 29, 8103–8120. [Google Scholar] [CrossRef] [PubMed]
  148. Dollé, L.; El Yazidi-Belkoura, I.; Adriaenssens, E.; Nurcombe, V.; Hondermarck, H. Nerve growth factor overexpression and autocrine loop in breast cancer cells. Oncogene 2003, 22, 5592–5601. [Google Scholar] [CrossRef]
  149. Lévêque, R.; Corbet, C.; Aubert, L.; Guilbert, M.; Lagadec, C.; Adriaenssens, E.; Duval, J.; Finetti, P.; Birnbaum, D.; Magné, N.; et al. ProNGF increases breast tumor aggressiveness through functional association of TrkA with EphA2. Cancer Lett. 2019, 449, 196–206. [Google Scholar] [CrossRef]
  150. Mazaki, A.; Yamauchi, K.; Orita, S.; Inage, K.; Suzuki, M.; Fujimoto, K.; Shiga, Y.; Abe, K.; Inoue, M.; Norimoto, M.; et al. Nerve Growth Factor in Breast Cancer Cells Promotes Axonal Growth and Expression of Calcitonin Gene-related Peptide in a Rat Model of Spinal Metastasis. Anticancer Res. 2022, 42, 581–587. [Google Scholar] [CrossRef]
  151. Gao, X.; Yang, Z.; Xu, C.; Yu, Q.; Wang, M.; Song, J.; Wu, C.; Chen, M. GeneChip expression profiling identified OLFML2A as a potential therapeutic target in TNBC cells. Ann. Transl. Med. 2022, 10, 274. [Google Scholar] [CrossRef]
  152. Di Donato, M.; Galasso, G.; Giovannelli, P.; Sinisi, A.A.; Migliaccio, A.; Castoria, G. Targeting the Nerve Growth Factor Signaling Impairs the Proliferative and Migratory Phenotype of Triple-Negative Breast Cancer Cells. Front. Cell Dev. Biol. 2021, 9, 676568. [Google Scholar] [CrossRef]
  153. Johansson, M.; Jonsson, M.; Norrgard, O.; Forsgren, S. New aspects concerning ulcerative colitis and colonic carcinoma: Analysis of levels of neuropeptides, neurotrophins, and TNFalpha/TNF receptor in plasma and mucosa in parallel with histological evaluation of the intestine. Inflamm. Bowel. Dis. 2008, 14, 1331–1340. [Google Scholar] [CrossRef] [PubMed]
  154. De Farias, C.; Stertz, L.; Lima, R.; Kapczinski, F.; Schwartsmann, G.; Roesler, R. Reduced NGF Secretion by HT-29 Human Colon Cancer Cells Treated with a GRPR Antagonist. Protein Pept. Lett. 2009, 16, 650–652. [Google Scholar] [CrossRef]
  155. Liebl, F.; Demir, I.E.; Rosenberg, R.; Boldis, A.; Yildiz, E.; Kujundzic, K.; Kehl, T.; Dischl, D.; Schuster, T.; Maak, M.; et al. The severity of neural invasion is associated with shortened survival in colon cancer. Clin. Cancer Res. 2013, 19, 50–61. [Google Scholar] [CrossRef] [PubMed]
  156. Jin, H.; Pan, Y.; He, L.; Zhai, H.; Li, X.; Zhao, L.; Sun, L.; Liu, J.; Hong, L.; Song, J.; et al. P75 Neurotrophin Receptor Inhibits Invasion and Metastasis of Gastric Cancer. Mol. Cancer Res. 2007, 5, 423–433. [Google Scholar] [CrossRef] [PubMed]
  157. Du, J.J.; Dou, K.F.; Peng, S.Y.; Qian, B.Z.; Xiao, H.S.; Liu, F.; Wang, W.Z.; Guan, W.X.; Gao, Z.Q.; Liu, Y.B.; et al. Expression of NGF family and their receptors in gastric carcinoma: A cDNA microarray study. World J. Gastroenterol. 2003, 9, 1431–1434. [Google Scholar] [CrossRef]
  158. Dou, N.; Yang, D.; Yu, S.; Wu, B.; Gao, Y.; Li, Y. SNRPA enhances tumour cell growth in gastric cancer through modulating NGF expression. Cell Prolif. 2018, 51, e12484. [Google Scholar] [CrossRef]
  159. Hayakawa, Y.; Sakitani, K.; Konishi, M.; Asfaha, S.; Niikura, R.; Tomita, H.; Renz, B.W.; Taylor, Y.; Macchini, M.; Middelholff, M.; et al. Nerve Growth Factor Promotes Gastric Tumorigenesis through Aberrant Cholinergic Signaling. Cancer Cell 2017, 31, 21–34. [Google Scholar] [CrossRef]
  160. Noh, S.J.; Kim, K.M.; Jang, K.Y. Individual and co-expression patterns of nerve growth factor and heme oxygenase-1 predict shorter survival of gastric carcinoma patients. Diagn. Pathol. 2017, 12, 48. [Google Scholar] [CrossRef]
  161. Dudás, J.; Dietl, W.; Romani, A.; Reinold, S.; Glueckert, R.; Schrott-Fischer, A.; Dejaco, D.; Chacko, L.J.; Tuertscher, R.; Schartinger, V.H.; et al. Nerve growth factor (NGF)—Receptor survival axis in head and neck squamous cell carcinoma. Int. J. Mol. Sci. 2018, 19, 1771. [Google Scholar] [CrossRef] [PubMed]
  162. Kolokythas, A.; Cox, D.P.; Dekker, N.; Schmidt, B.L. Nerve Growth Factor and Tyrosine Kinase A Receptor in Oral Squamous Cell Carcinoma: Is There an Association with Perineural Invasion? J. Oral Maxillofac. Surg. 2010, 68, 1290–1295. [Google Scholar] [CrossRef] [PubMed]
  163. Wang, L.; Sun, M.; Jiang, Y.; Yang, L.; Lei, D.; Lu, C.; Zhao, Y.; Zhang, P.; Yang, Y.; Li, J. Nerve growth factor and tyrosine kinase A in human salivary adenoid cystic carcinoma: Expression patterns and effects on in vitro invasive behavior. J. Oral. Maxillofac. Surg. 2006, 64, 636–641. [Google Scholar] [CrossRef] [PubMed]
  164. Assimakopoulou, M.; Zolota, V.; Chondrogianni, C.; Gatzounis, G.; Varakis, J. p75NTR and TrkC neurotrophin receptors demonstrate a different immunoreactivity profile in comparison to TrkA and TrkB receptors in human normal pituitary gland and adenomas. Neuroendocrinology 2008, 88, 127–134. [Google Scholar] [CrossRef] [PubMed]
  165. Faulkner, S.; Roselli, S.; Demont, Y.; Pundavela, J.; Choquet, G.; Leissner, P.; Oldmeadow, C.; Attia, J.; Walker, M.M.; Hondermarck, H. ProNGF is a potential diagnostic biomarker for thyroid cancer. Oncotarget 2016, 7, 28488–28497. [Google Scholar] [CrossRef]
  166. Griffin, N.; Gao, F.; Jobling, P.; Oldmeadow, C.; Wills, V.; Walker, M.M.; Faulkner, S.; Hondermarck, H. The neurotrophic tyrosine kinase receptor 1 (TrkA) is overexpressed in oesophageal squamous cell carcinoma. Pathology 2021, 53, 470–477. [Google Scholar] [CrossRef]
  167. Ye, Y.; Dang, D.; Zhang, J.; Viet, C.T.; Lam, D.K.; Dolan, J.C.; Gibbs, J.L.; Schmidt, B.L. Nerve growth factor links oral cancer progression, pain, and cachexia. Mol. Cancer Ther. 2011, 10, 1667–1676. [Google Scholar] [CrossRef]
  168. Søland, T.M.; Brusevold, I.J.; Koppang, H.S.; Schenck, K.; Bryne, M. Nerve growth factor receptor (p75NTR) and pattern of invasion predict poor prognosis in oral squamous cell carcinoma. Histopathology 2008, 53, 62–72. [Google Scholar] [CrossRef]
  169. Alzawi, A.; Iftikhar, A.; Shalgm, B.; Jones, S.; Ellis, I.; Islam, M. Receptor, Signal, Nucleus, Action: Signals That Pass through Akt on the Road to Head and Neck Cancer Cell Migration. Cancers 2022, 14, 2606. [Google Scholar] [CrossRef]
  170. Herbrich, S.M.; Kannan, S.; Nolo, R.M.; Hornbaker, M.; Chandra, J.; Zweidler-McKay, P.A. Characterization of TRKA signaling in acute myeloid leukemia. Oncotarget 2018, 9, 30092–30105. [Google Scholar] [CrossRef]
  171. Lebedev, T.D.; Vagapova, E.R.; Popenko, V.I.; Leonova, O.G.; Spirin, P.V.; Prassolov, V.S. Two receptors, two isoforms, two cancers: Comprehensive analysis of kit and trka expression in neuroblastoma and acute myeloid leukemia. Front. Oncol. 2019, 9, 1046. [Google Scholar] [CrossRef] [PubMed]
  172. Hillis, J.; O’Dwyer, M.; Gorman, A.M. Neurotrophins and B-cell malignancies. Cell Mol. Life Sci. 2016, 73, 41–56. [Google Scholar] [CrossRef] [PubMed]
  173. Franzese, O.; Di Francesco, A.M.; Meco, D.; Graziani, G.; Cusano, G.; Levati, L.; Riccardi, R.; Ruggiero, A. hTERT transduction extends the lifespan of primary pediatric low-grade glioma cells while preserving the biological response to NGF. Pathol. Oncol. Res. 2021, 27, 612375. [Google Scholar] [CrossRef] [PubMed]
  174. Damm, R.; Pech, M.; Cavalli, P.; Haag, F.; Gylstorff, S.; Omari, J.; Thormann, M.; Seidensticker, R.; Ricke, J.; Seidensticker, M.; et al. Correlation of chemokines and growth factors with radiation-induced liver injury after interstitial high dose rate (HDR) brachytherapy of liver metastases. J. Cancer Res. Clin. Oncol. 2022, 148, 2815–2826. [Google Scholar] [CrossRef] [PubMed]
  175. Kishibe, K.; Yamada, Y.; Ogawa, K. Production of nerve growth factor by mouse hepatocellular carcinoma cells and expression of TrkA in tumor-associated arteries in mice. Gastroenterology 2002, 122, 1978–1986. [Google Scholar] [CrossRef] [PubMed]
  176. Koizumi, H.; Morita, M.; Mikami, S.; Shibayama, E.; Uchikoshi, T. Immunohistochemical analysis of TrkA neurotrophin receptor expression in human non-neuronal carcinomas. Pathol. Int. 1998, 48, 93–101. [Google Scholar] [CrossRef] [PubMed]
  177. Trim, N.; Morgan, S.; Evans, M.; Issa, R.; Fine, D.; Afford, S.; Winkins, B.; Iredale, J. Hepatic stellate cells express the low affinity nerve growth factor receptor p75 and undergo apoptosis in response to nerve growth factor stimulation. Am. J. Pathol. 2000, 156, 1235–1243. [Google Scholar] [CrossRef]
  178. Rasi, G.; Serafino, A.; Bellis, L.; Lonardo, M.T.; Andreola, F.; Zonfrillo, M.; Vennarecci, G.; Pierimarchi, P.; Sinibaldi Vallebona, P.; Ettorre, G.M.; et al. Nerve growth factor involvement in liver cirrhosis and hepatocellular carcinoma. World J. Gastroenterol. 2007, 13, 4986–4995. [Google Scholar] [CrossRef]
  179. Malaguarnera, G.; Giordano, M.; Paladina, I.; Berretta, M.; Cappellani, A.; Malaguarnera, M. Serum markers of hepatocellular carcinoma. Dig. Dis. Sci. 2010, 55, 2744–2755. [Google Scholar] [CrossRef]
  180. Luo, T.; Zhang, S.G.; Zhu, L.F.; Zhang, F.X.; Li, W.; Zhao, K.; Wen, X.X.; Yu, M.; Zhan, Y.K.; Chen, H.; et al. A selective c-Met and Trks inhibitor Indo5 suppresses hepatocellular carcinoma growth. J. Exp. Clin. Cancer Res. 2019, 38, 130. [Google Scholar] [CrossRef]
  181. Natri, H.M.; Wilson, M.A.; Buetow, K.H. Distinct molecular etiologies of male and female hepatocellular carcinoma. BMC Cancer 2019, 19, 951. [Google Scholar] [CrossRef] [PubMed]
  182. Sherif, I.O.; Al-Gayyar, M.M.H. Oleuropein potentiates anti-tumor activity of cisplatin against HepG2 through affecting proNGF/NGF balance. Life Sci. 2018, 198, 87–93. [Google Scholar] [CrossRef] [PubMed]
  183. Thompson, S.M.; Jondal, D.E.; Butters, K.A.; Knudsen, B.E.; Anderson, J.L.; Roberts, L.R.; Callstrom, M.R.; Woodrum, D.A. Heat stress and thermal ablation induce local expression of nerve growth factor inducible (VGF) in hepatocytes and hepatocellular carcinoma: Preclinical and clinical studies. Gene Expr. 2018, 19, 37–47. [Google Scholar] [CrossRef]
  184. Tokusashi, Y.; Asai, K.; Tamakawa, S.; Yamamoto, M.; Yoshie, M.; Yaginuma, Y.; Miyokawa, N.; Aoki, T.; Kino, S.; Kasai, S.; et al. Expression of NGF in hepatocellular carcinoma cells with its receptors in non-tumor cell components. Int. J. Cancer 2005, 114, 39–45. [Google Scholar] [CrossRef] [PubMed]
  185. Owusu Sekyere, S.; Port, K.; Deterding, K.; Cornberg, M.; Wedemeyer, H. Inflammatory patterns in plasma associate with hepatocellular carcinoma development in cured hepatitis C cirrhotic patients. United Eur. Gastroenterol. J. 2021, 9, 486–496. [Google Scholar] [CrossRef]
  186. Ye, L.; Wu, Y.; Zhou, J.; Xie, M.; Zhang, Z.; Su, C. Influence of Exosomes on Astrocytes in the Pre-Metastatic Niche of Lung Cancer Brain Metastases. Biol. Proced. Online 2023, 25, 5. [Google Scholar] [CrossRef]
  187. Ricci, A.; Salvucci, C.; Castelli, S.; Carraturo, A.; de Vitis, C.; D’Ascanio, M. Adenocarcinomas of the Lung and Neurotrophin System: A Review. Biomedicines 2022, 10, 2531. [Google Scholar] [CrossRef]
  188. Gao, F.; Griffin, N.; Faulkner, S.; Rowe, C.W.; Williams, L.; Roselli, S.; Thorne, R.F.; Ferdoshi, A.; Jobling, P.; Walker, M.M.; et al. The neurotrophic tyrosine kinase receptor TrkA and its ligand NGF are increased in squamous cell carcinomas of the lung. Sci. Rep. 2018, 8, 8135. [Google Scholar] [CrossRef]
  189. Vaishnavi, A.; Capelletti, M.; Le, A.T.; Kako, S.; Butaney, M.; Ercan, D.; Mahale, S.; Davies, K.D.; Aisner, D.L.; Pilling, A.B.; et al. Oncogenic and drug-sensitive NTRK1 rearrangements in lung cancer. Nat. Med. 2013, 19, 1469–1472. [Google Scholar] [CrossRef]
  190. Jiang, Q.; Li, M.; Li, H.; Chen, L. Entrectinib, a new multi-target inhibitor for cancer therapy. Biomed. Pharmacother. 2022, 150, 112974. [Google Scholar] [CrossRef]
  191. Ricci, A.; Greco, S.; Mariotta, S.; Felici, L.; Bronzetti, E.; Cavazzana, A.; Cardillo, G.; Amenta, F.; Bisetti, A.; Barbolini, G. Neurotrophins and neurotrophin receptors in human lung cancer. Am. J. Respir. Cell Mol. Biol. 2001, 25, 439–446. [Google Scholar] [CrossRef] [PubMed]
  192. Retamales-Ortega, R.; Oróstica, L.; Vera, C.; Cuevas, P.; Hernández, A.; Hurtado, I.; Vega, M.; Romero, C. Role of nerve growth factor (NGF) and miRNAs in epithelial ovarian cancer. Int. J. Mol. Sci. 2017, 18, 507. [Google Scholar] [CrossRef] [PubMed]
  193. Campos, X.; Muñoz, Y.; Selman, A.; Yazigi, R.; Moyano, L.; Weinstein-Oppenheimer, C.; Lara, H.E.; Romero, C. Nerve growth factor and its high-affinity receptor trkA participate in the control of vascular endothelial growth factor expression in epithelial ovarian cancer. Gynecol. Oncol. 2007, 104, 168–175. [Google Scholar] [CrossRef] [PubMed]
  194. Vera, D.B.; Fredes, A.N.; Garrido, M.P.; Romero, C. Role of mitochondria in interplay between ngf/trka, mir-145 and possible therapeutic strategies for epithelial ovarian cancer. Life 2022, 12, 8. [Google Scholar] [CrossRef]
  195. Garrido, M.P.; Salvatierra, R.; Valenzuela-Valderrama, M.; Vallejos, C.; Bruneau, N.; Hernández, A.; Vega, M.; Selman, A.; Quest, A.F.G.; Romero, C. Metformin reduces ngf-induced tumour promoter effects in epithelial ovarian cancer cells. Pharmaceuticals 2020, 13, 315. [Google Scholar] [CrossRef]
  196. Garrido, M.P.; Torres, I.; Avila, A.; Chnaiderman, J.; Valenzuela-Valderrama, M.; Aramburo, J.; Orostica, L.; Duran-Jara, E.; Lobos-Gonzalez, L.; Romero, C. Ngf/trka decrease mir-145-5p levels in epithelial ovarian cancer cells. Int. J. Mol. Sci. 2020, 21, 7657. [Google Scholar] [CrossRef]
  197. Wijaya, L.K.; Stumbles, P.A.; Drummond, P.D. A positive feedback loop between alpha1-adrenoceptors and inflammatory cytokines in keratinocytes. Exp. Cell Res. 2020, 391, 112008. [Google Scholar] [CrossRef]
  198. Tapia, V.; Gabler, F.; Muñoz, M.; Yazigi, R.; Paredes, A.; Selman, A.; Vega, M.; Romero, C. Tyrosine kinase A receptor (trkA): A potential marker in epithelial ovarian cancer. Gynecol. Oncol. 2011, 121, 13–23. [Google Scholar] [CrossRef]
  199. Yu, X.; Liu, Z.; Hou, R.; Nie, Y.; Chen, R. Nerve growth factor and its receptors on onset and diagnosis of ovarian cancer. Oncol. Lett. 2017, 14, 2864–2868. [Google Scholar] [CrossRef]
  200. Brodeur, G.M.; Minturn, J.E.; Ho, R.; Simpson, A.M.; Iyer, R.; Varela, C.R.; Light, J.E.; Kolla, V.; Evans, A.E. Trk receptor expression and inhibition in neuroblastomas. Clin. Cancer Res. 2009, 15, 3244–3250. [Google Scholar] [CrossRef]
  201. Hoehner, J.C.; Olsen, L.; Sandstedt, B.; Kaplan, D.R.; Pahlman, S. Association of neurotrophin receptor expression and differentiation in human neuroblastoma. Am. J. Pathol. 1995, 147, 102–113. [Google Scholar]
  202. Nakagawara, A.; Arima, M.; Azar, C.G.; Scavarda, N.J.; Brodeur, G.M. Inverse Relationship between trk Expression and N-myc Amplification in Human Neuroblastomas. Cancer Res. 1992, 52, 1364–1368. [Google Scholar]
  203. Dzieran, J.; Garcia, A.R.; Westermark, U.K.; Henley, A.B.; Sánchez, E.E.; Träger, C.; Johansson, H.J.; Lehtiö, J.; Arsenian-Henriksson, M. MYCN-amplified neuroblastoma maintains an aggressive and undifferentiated phenotype by deregulation of estrogen and NGF signaling. Proc. Natl. Acad. Sci. USA 2018, 115, E1229–E1238. [Google Scholar] [CrossRef]
  204. Eggert, A.; Ikegaki, N.; Liu, X.G.; Brodeur, G.M. Prognostic and biological role of neurotrophin- receptor TrkA and TrkB in neuroblastoma. Klin. Padiatr. 2000, 212, 200–205. [Google Scholar] [CrossRef]
  205. Ho, R.; Minturn, J.E.; Simpson, A.M.; Iyer, R.; Light, J.E.; Evans, A.E.; Brodeur, G.M. The effect of P75 on Trk receptors in neuroblastomas. Cancer Lett. 2011, 305, 76–85. [Google Scholar] [CrossRef]
  206. Cai, S.; Chen, Q.; Xu, Y.; Zhuang, Q.; Ji, S. Atorvastatin inhibits pancreatic cancer cells proliferation and invasion likely by suppressing neurotrophin receptor signaling. Transl. Cancer Res. 2020, 9, 1439–1447. [Google Scholar] [CrossRef]
  207. Bapat, A.A.; Munoz, R.M.; Von Hoff, D.D.; Han, H. Blocking nerve growth factor signaling reduces the neural invasion potential of pancreatic cancer cells. PLoS ONE 2016, 11, e0165586. [Google Scholar] [CrossRef]
  208. Lei, Y.; Tang, L.; Xie, Y.; Xianyu, Y.; Zhang, L.; Wang, P.; Hamada, Y.; Jiang, K.; Zheng, W.; Jiang, X. Gold nanoclusters-assisted delivery of NGF siRNA for effective treatment of pancreatic cancer. Nat. Commun. 2017, 8, 15130. [Google Scholar] [CrossRef]
  209. Xu, J.B.; Yang, X.C.; Guo, J.H.; Chen, J.Y.; Shang, Y.; Liu, W.J.; Wang, T.-M.; Zou, L. Serum nerve growth factor level indicates therapeutic efficacy of 125I seed implantation in advanced pancreatic adenocarcinoma. Eur. Rev. Med. Pharmacol. Sci. 2015, 19, 3385–3390. [Google Scholar]
  210. Zhang, Y.; Dang, C.; Ma, Q.; Shimahara, Y. Expression of nerve growth factor receptors and their prognostic value in human pancreatic cancer. Oncol. Rep. 2005, 14, 161–171. [Google Scholar]
  211. Dang, C.; Zhang, Y.; Ma, Q.; Shimahara, Y. Expression of nerve growth factor receptors is correlated with progression and prognosis of human pancreatic cancer. J. Gastroenterol. Hepatol. 2006, 21, 850–858. [Google Scholar] [CrossRef]
  212. Ma, J.; Jiang, Y.; Jiang, Y.; Sun, Y.; Zhao, X. Expression of nerve growth factor and tyrosine kinase receptor a and correlation with perineural invasion in pancreatic cancer. J. Gastroenterol. Hepatol. 2008, 23, 1852–1859. [Google Scholar] [CrossRef]
  213. Lanki, M.; Mustonen, H.; Salmi, M.; Jalkanen, S.; Haglund, C.; Seppänen, H. Serum cytokine profiles in patients with pancreatic cancer and chronic pancreatitis. Pancreatology 2023, 23, 657–662. [Google Scholar] [CrossRef]
  214. Qin, T.; Xiao, Y.; Qian, W.; Wang, X.; Gong, M.; Wang, Q.; An, R.; Han, L.; Duan, W.; Ma, Q.; et al. HGF/c-Met pathway facilitates the perineural invasion of pancreatic cancer by activating the mTOR/NGF axis. Cell Death Dis. 2022, 13, 387. [Google Scholar] [CrossRef]
  215. Baker, D.L.; Molenaar, W.M.; Trojanowski, J.Q.; Evans, A.E.; Ross, A.H.; Rorke, L.B.; Parker, R.J.; Lee, V.M.; Pleasure, D. Nerve growth factor receptor expression in peripheral and central neuroectodermal tumors, other pediatric brain tumors, and during development of the adrenal gland. Am. J. Pathol. 1991, 139, 115–122. [Google Scholar]
  216. Falsini, B.; Chiaretti, A.; Barone, G.; Piccardi, M.; Pierri, F.; Colosimo, C.; Lazzareschi, I.; Ruggiero, A.; Parisi, V.; Fadda, A.; et al. Topical nerve growth factor as a visual rescue strategy in pediatric optic gliomas: A pilot study including electrophysiology. Neurorehabil. Neural. Repair. 2011, 25, 512–520. [Google Scholar] [CrossRef]
  217. Al-Ghazzawi, K.; Wessolly, M.; Dalbah, S.; Ketteler, P.; Kiefer, T.; Bechrakis, N.; Leyla, J.; Ting, S.; Biewald, E.; Mairinger, F.D. PDGF, NGF, and EGF as main contributors to tumorigenesis in high-risk retinoblastoma. Front. Oncol. 2023, 13, 1144951. [Google Scholar] [CrossRef]
  218. Shen, T.; Li, Y.; Wang, D.; Su, Y.; Li, G.; Shang, Z.; Niu, J.; Tan, X. YAP1-TEAD1 mediates the perineural invasion of prostate cancer cells induced by cancer-associated fibroblasts. Biochim. Biophys. Acta—Mol. Basis Dis. 2022, 1868, 166540. [Google Scholar] [CrossRef]
  219. Di Donato, M.; Giovannelli, P.; Migliaccio, A.; Castoria, G. The nerve growth factor-delivered signals in prostate cancer and its associated microenvironment: When the dialogue replaces the monologue. Cell Biosci. 2023, 13, 60. [Google Scholar] [CrossRef]
  220. Zhang, Y.; Li, W.; Guo, S.; Wu, Z.; Zhang, L.; Liu, Y.; Li, X.; Guo, X.; Cao, J.; Yang, C.; et al. FBXO22 Mediates the NGF/TRKA Signaling Pathway in Bone Metastases in Prostate Cancer. Am. J. Pathol. 2023, 193, 1248–1266. [Google Scholar] [CrossRef]
  221. Di Donato, M.; Cernera, G.; Auricchio, F.; Migliaccio, A.; Castoria, G. Cross-talk between androgen receptor and nerve growth factor receptor in prostate cancer cells: Implications for a new therapeutic approach. Cell Death Discov. 2018, 4, 5. [Google Scholar] [CrossRef]
  222. Pflug, B.; Djakiew, D. Expression of p75(NTR) in a human prostate epithelial tumor cell line reduces nerve growth factor-induced cell growth by activation of programmed cell death. Mol. Carcinog. 1998, 23, 106–114. [Google Scholar] [CrossRef]
  223. Pundavela, J.; Demont, Y.; Jobling, P.; Lincz, L.F.; Roselli, S.; Thorne, R.F.; Bond, D.; Bradshaw, R.A.; Walker, M.M.; Hondermarck, H. ProNGF correlates with Gleason score and is a potential driver of nerve infiltration in prostate cancer. Am. J. Pathol. 2014, 184, 3156–3162. [Google Scholar] [CrossRef]
  224. Chen-Tsai, C.P.; Colome-Grimmer, M.; Wagner, R.F. Correlations among neural cell adhesion molecule, nerve growth factor, and its receptors, TrkA, TrkB, TrkC, and p75NGFR, in perineural invasion by basal cell and cutaneous squamous cell carcinomas. Dermatol. Surg. 2004, 30, 1009–1016. [Google Scholar] [CrossRef]
  225. Pasini, L.; Re, A.; Tebaldi, T.; Ricci, G.; Boi, S.; Adami, V.; Barbareschi, M.; Quattrone, A. TrkA is amplified in malignant melanoma patients and induces an anti-proliferative response in cell lines. BMC Cancer 2015, 15, 777. [Google Scholar] [CrossRef]
  226. Chan, M.M.; Tahan, S.R. Low-affinity nerve growth factor receptor (P75 NGFR) as a marker of perineural invasion in malignant melanomas. J. Cutan. Pathol. 2010, 37, 336–343. [Google Scholar] [CrossRef]
  227. Wehkamp, U.; Stern, S.; Krüger, S.; Weichenthal, M.; Hauschild, A.; Röcken, C.; Egberts, F. Co-expression of NGF and PD-L1 on tumor-associated immune cells in the microenvironment of Merkel cell carcinoma. J. Cancer Res. Clin. Oncol. 2018, 144, 1301–1308. [Google Scholar] [CrossRef]
  228. Bröcker, E.B.; Magiera, H.; Herlyn, M. Nerve growth and expression of receptors for nerve growth factor in tumors of melanocyte origin. J. Investig. Dermatol. 1991, 96, 662–665. [Google Scholar] [CrossRef]
  229. Mirkina, I.; Hadzijusufovic, E.; Krepler, C.; Mikula, M.; Mechtcheriakova, D.; Strommer, S.; Stella, A.; Jensen-Jarolim, E.; Holler, C.; Wacheck, V.; et al. Phenotyping of human melanoma cells reveals a unique composition of receptor targets and a subpopulation co-expressing ErbB4, EPO-R and NGF-R. PLoS ONE 2014, 9, e84417. [Google Scholar] [CrossRef]
  230. Innominato, P.F.; Libbrecht, L.; van den Oord, J.J. Expression of neurotrophins and their receptors in pigment cell lesions of the skin. J. Pathol. 2001, 194, 95–100. [Google Scholar] [CrossRef]
  231. Sigal, A.C.; Keenan, M.; Lazova, R. P75 nerve growth factor receptor as a useful marker to distinguish spindle cell melanoma from other spindle cell neoplasms of sun-damaged skin. Am. J. Dermatopathol. 2012, 34, 145–150. [Google Scholar] [CrossRef]
  232. Lazova, R.; Tantcheva-Poor, I.; Sigal, A.C. P75 nerve growth factor receptor staining is superior to S100 in identifying spindle cell and desmoplastic melanoma. J. Am. Acad. Dermatol. 2010, 63, 852–858. [Google Scholar] [CrossRef]
  233. Truzzi, F.; Marconi, A.; Lotti, R.; Dallaglio, K.; French, L.E.; Hempstead, B.L.; Pincelli, C. Neurotrophins and their receptors stimulate melanoma cell proliferation and migration. J. Investig. Dermatol. 2008, 128, 2031–2040. [Google Scholar] [CrossRef]
  234. Miao, Q.; Ma, K.; Chen, D.; Wu, X.; Jiang, S. Targeting tropomyosin receptor kinase for cancer therapy. Eur. J. Med. Chem. 2019, 175, 129–148. [Google Scholar] [CrossRef]
  235. Ardini, E.; Bosotti, R.; Borgia, A.L.; De Ponti, C.; Somaschini, A.; Cammarota, R.; Amboldi, N.; Raddrizzani, L.; Milani, A.; Magnaghi, P.; et al. The TPM3-NTRK1 rearrangement is a recurring event in colorectal carcinoma and is associated with tumor sensitivity to TRKA kinase inhibition. Mol. Oncol. 2014, 8, 1495–1507. [Google Scholar] [CrossRef]
  236. Pinto, A.; Nosé, V.; Rojas, C.; Fan, Y.S.; Gomez-Fernandez, C. Searching for mammary analog secretory carcinoma of salivary gland among its mimics. Mod. Pathol. 2014, 27, 30–37. [Google Scholar] [CrossRef]
  237. Venkat, S.; Fitzpatrick, S.; Drew, P.A.; Bhattacharyya, I.; Cohen, D.M.; Islam, M.N. Secretory Carcinoma of the Oral Cavity: A Retrospective Case Series with Review of Literature. Head Neck Pathol. 2021, 15, 893–904. [Google Scholar] [CrossRef]
  238. Morerio, C.; Rapella, A.; Rosanda, C.; Tassano, E.; Conte, M.; Gambini, C.; Panarello, C. Differential diagnosis of congenital fibrosarcoma. Cancer Genet. Cytogenet. 2004, 152, 167–168. [Google Scholar] [CrossRef]
  239. Eguchi, M.; Eguchi-Ishimae, M.; Tojo, A.; Morishita, K.; Suzuki, K.; Sato, Y.; Kudoh, S.; Tanaka, K.; Setoyama, M.; Nagamura, F.; et al. Fusion of ETV6 to neurotrophin-3 receptor TRKC in acute myeloid leukemia with t(12;15)(p13;q25). Blood 1999, 93, 1355–1363. [Google Scholar] [CrossRef]
  240. Eguchi, M.; Eguchi-Ishimae, M. Letter to the editor: Absence of t(12;15) associated ETV6-NTRK3 fusion transcripts in pediatric acute leukemias. Med. Pediatr. Oncol. 2001, 37, 417. [Google Scholar] [CrossRef]
  241. Créancier, L.; Vandenberghe, I.; Gomes, B.; Dejean, C.; Blanchet, J.C.; Meilleroux, J.; Guimbaud, R.; Selves, J.; Kruczynski, A. Chromosomal rearrangements involving the NTRK1 gene in colorectal carcinoma. Cancer Lett. 2015, 365, 107–111. [Google Scholar] [CrossRef]
  242. Bourgeois, J.M.; Knezevich, S.R.; Mathers, J.A.; Sorensen, P.H.B. Molecular detection of the ETV6-NTRK3 gene fusion differentiates congenital fibrosarcoma from other childhood spindle cell tumors. Am. J. Surg. Pathol. 2000, 24, 937–946. [Google Scholar] [CrossRef]
  243. Alam, M.S.; Choi, S.U.; Lee, D.U. Synthesis, anticancer, and docking studies of salicyl-hydrazone analogues: A novel series of small potent tropomyosin receptor kinase A inhibitors. Bioorganic Med. Chem. 2017, 25, 389–396. [Google Scholar] [CrossRef]
  244. Malekan, M.; Nezamabadi, S.S.; Samami, E.; Mohebalizadeh, M.; Saghazadeh, A.; Rezaei, N. BDNF and its signaling in cancer. J. Cancer Res. Clin. Oncol. 2023, 149, 2621–2636. [Google Scholar] [CrossRef]
  245. Moore, L.D.; Le, T.; Fan, G. DNA methylation and its basic function. Neuropsychopharmacology 2013, 38, 23–38. [Google Scholar] [CrossRef]
  246. Zhu, X.; Chen, Z.; Shen, W.; Huang, G.; Sedivy, J.M.; Wang, H.; Ju, Z. Inflammation, epigenetics, and metabolism converge to cell senescence and ageing: The regulation and intervention. Signal Transduct. Target Ther. 2021, 6, 245. [Google Scholar] [CrossRef]
  247. Fiore, M.; Messina, M.P.; Petrella, C.; D’Angelo, A.; Greco, A.; Ralli, M.; Ferraguti, G.; Tarani, L.; Ceccanti, M. Antioxidant properties of plant polyphenols in the counteraction of alcohol-abuse induced damage: Impact on the Mediterranean diet. J. Funct. Foods 2020, 71, 104012. [Google Scholar] [CrossRef]
  248. Gasche, J.A.; Hoffmann, J.; Boland, C.R.; Goel, A. Interleukin-6 promotes tumorigenesis by altering DNA methylation in oral cancer cells. Int. J. Cancer 2011, 129, 1053–1063. [Google Scholar] [CrossRef]
  249. Ray, D.; Yung, R. Immune senescence, epigenetics and autoimmunity. Clin. Immunol. 2018, 196, 59–63. [Google Scholar] [CrossRef]
  250. Petrella, C.; Di Certo, M.G.; Gabanella, F.; Barbato, C.; Ceci, F.M.; Greco, A.; Ralli, M.; Polimeni, A.; Angeloni, A.; Severini, C.; et al. Mediterranean Diet, Brain and Muscle: Olive Polyphenols and Resveratrol Protection in Neurodegenerative and Neuromuscular Disorders. Curr. Med. Chem. 2021, 28, 7595–7613. [Google Scholar] [CrossRef]
  251. Zhang, L.; Lu, Q.; Chang, C. Epigenetics in Health and Disease. Adv. Exp. Med. Biol. 2020, 1253, 3–55. [Google Scholar] [CrossRef] [PubMed]
  252. Irimie, A.I.; Ciocan, C.; Gulei, D.; Mehterov, N.; Atanasov, A.G.; Dudea, D.; Berindan-Neagoe, I. Current insights into oral cancer epigenetics. Int. J. Mol. Sci. 2018, 19, 670. [Google Scholar] [CrossRef] [PubMed]
  253. Colucci-D’amato, L.; Speranza, L.; Volpicelli, F. Neurotrophic factor bdnf, physiological functions and therapeutic potential in depression, neurodegeneration and brain cancer. Int. J. Mol. Sci. 2020, 21, 7777. [Google Scholar] [CrossRef]
  254. Zhang, M.; Zheng, H.; Zhang, X.; Tian, X.; Xu, S.; Liu, Y.; Jiang, S.; Liu, X.; Shi, R.; Gong, K.; et al. Involvement of nerve growth factor in mouse hippocampal neuronal cell line (HT22) differentiation and underlying role of DNA methyltransferases. J. Toxicol. Environ. Health Part A 2018, 81, 1116–1122. [Google Scholar] [CrossRef]
  255. Yuan, H.; Du, S.; Chen, L.; Xu, X.; Wang, Y.; Ji, F. Hypomethylation of nerve growth factor (NGF) promotes binding of C/EBPα and contributes to inflammatory hyperalgesia in rats. J. Neuroinflamm. 2020, 17, 34. [Google Scholar] [CrossRef]
  256. Balakrishnan, A.; Guruprasad, K.P.; Satyamoorthy, K.; Joshi, M.B. Interleukin-6 determines protein stabilization of DNA methyltransferases and alters DNA promoter methylation of genes associated with insulin signaling and angiogenesis. Lab. Investig. 2018, 98, 1143–1158. [Google Scholar] [CrossRef]
  257. Lian, B.S.X.; Kawasaki, T.; Kano, N.; Ori, D.; Ikegawa, M.; Isotani, A.; Kawai, T. Regulation of Il6 expression by single CpG methylation in downstream of Il6 transcription initiation site. IScience 2022, 25, 104118. [Google Scholar] [CrossRef]
  258. Jin, H.; Wu, Z.; Tan, B.; Liu, Z.; Zu, Z.; Wu, X.; Bi, Y.; Hu, X. Ibuprofen promotes p75 neurotrophin receptor expression through modifying promoter methylation and N6-methyladenosine-RNA-methylation in human gastric cancer cells. Bioengineered 2022, 13, 14595–14604. [Google Scholar] [CrossRef] [PubMed]
  259. Terracina, S.; Ferraguti, G.; Tarani, L.; Messina, M.P.; Lucarelli, M.; Vitali, M.; De Persis, S.; Greco, A.; Minni, A.; Polimeni, A.; et al. Transgenerational Abnormalities Induced by Paternal Preconceptual Alcohol Drinking. Findings from Humans and Animal Models. Curr. Neuropharmacol. 2021, 19, 1158–1173. [Google Scholar] [CrossRef]
  260. Daskalakis, M.; Brocks, D.; Sheng, Y.H.; Islam, M.S.; Ressnerova, A.; Assenov, Y.; Milde, T.; Oehme, I.; Witt, O.; Goyal, A.; et al. Reactivation of endogenous retroviral elements via treatment with DNMT-and HDAC-inhibitors. Cell Cycle 2018, 17, 811–822. [Google Scholar] [CrossRef]
  261. Bellanger, C.; Dubanet, L.; Lise, M.C.; Fauchais, A.L.; Bordessoule, D.; Jauberteau, M.O.; Troutaud, D. Endogenous neurotrophins and Trk signaling in diffuse large B cell lymphoma cell lines are involved in sensitivity to rituximab-induced apoptosis. PLoS ONE 2011, 6, e27213. [Google Scholar] [CrossRef] [PubMed]
  262. Krytska, K.; Ryles, H.T.; Sano, R.; Raman, P.; Infarinato, N.R.; Hansel, T.D.; Makena, M.R.; Song, M.M.; Patrick Reynolds, C.; Mossé, Y.P. Crizotinib synergizes with chemotherapy in preclinical models of neuroblastoma. Clin. Cancer Res. 2016, 22, 948–960. [Google Scholar] [CrossRef] [PubMed]
  263. Pacenta, H.L.; Macy, M.E. Entrectinib and other ALK/TRK inhibitors for the treatment of neuroblastoma. Drug. Des. Dev. Ther. 2018, 12, 3549–3561. [Google Scholar] [CrossRef] [PubMed]
  264. Liang, Z.; Tan, J.; Tang, L.; Xie, Z.; Chen, G.; Liu, G.; Yuan, L.; Wang, K.-X.; Ding, H.-P.; Qiu, H.; et al. NGF monoclonal antibody DS002 alleviates chemotherapy-induced peripheral neuropathy in rats. Acta Pharmacol. Sin. 2022, 43, 2841–2847. [Google Scholar] [CrossRef]
  265. Cocco, E.; Scaltriti, M.; Drilon, A. NTRK fusion-positive cancers and TRK inhibitor therapy. Nat. Rev. Clin. Oncol. 2018, 15, 731–747. [Google Scholar] [CrossRef]
Figure 1. Role of chronic inflammation in cancer. Chronic inflammation originating from persistent stimuli has been associated with many steps of the carcinogenesis process including transformation, promotion, survival, proliferation, invasion, angiogenesis, and metastasis. (1) Cellular transformation is favored by the mutagenic action of ROS released by the immune cells and the action of TNF-α released by macrophages and T lymphocytes. (2) Carcinogenesis is promoted by various cytokines released during the chronic inflammatory process, including IL-1, IL-6, and TNF-α released by macrophages. (3) The survival of the tumor is associated with an ineffective response of the immune system associated with the defective action of inflammatory cells (like the release of IL-4 and IL-5 by T cells associated with T-helper 2 but not T-helper 1 responses) related to the release of various molecules like TNF-α, VEGF, Fas ligand, and transforming growth factor-β. (4) The invasion is favored by numerous molecules during inflammatory states; some of the most important are those associated with hypoxia including HIF, TNF-α, IL-1, and IL-6. (5) Inflammatory cells, especially macrophages, but also endothelial cells and platelets stimulate vascular growth through the release of many angiogenic factors (e.g., VEGF, IL-8, FGF, PDGF), sustaining the nutritional needs of the tumor and favoring its survival and migration. (6) Numerous cytokines and factors released during inflammation can lead to metastatic events of tumors including IL-6 and IL-10. The inflammatory microenvironment has a major role in this setting with implications for the prevention and treatment of cancer. FGF, fibroblast growth factor; HIF, hypoxia-inducible factor; IL, interleukin; PDGF, platelet-derived growth factor; ROS, reactive oxygen species; VEGF, vascular endothelial growth factor; TNF-α, tumor necrosis factor. Parts of the figure were drawn by using pictures from Servier Medical Art. Servier Medical Art by Servier is licensed under a Creative Commons Attribution 3.0 Unported License (https://creativecommons.org/licenses/by/4.0/, accessed on 21 January 2024).
Figure 1. Role of chronic inflammation in cancer. Chronic inflammation originating from persistent stimuli has been associated with many steps of the carcinogenesis process including transformation, promotion, survival, proliferation, invasion, angiogenesis, and metastasis. (1) Cellular transformation is favored by the mutagenic action of ROS released by the immune cells and the action of TNF-α released by macrophages and T lymphocytes. (2) Carcinogenesis is promoted by various cytokines released during the chronic inflammatory process, including IL-1, IL-6, and TNF-α released by macrophages. (3) The survival of the tumor is associated with an ineffective response of the immune system associated with the defective action of inflammatory cells (like the release of IL-4 and IL-5 by T cells associated with T-helper 2 but not T-helper 1 responses) related to the release of various molecules like TNF-α, VEGF, Fas ligand, and transforming growth factor-β. (4) The invasion is favored by numerous molecules during inflammatory states; some of the most important are those associated with hypoxia including HIF, TNF-α, IL-1, and IL-6. (5) Inflammatory cells, especially macrophages, but also endothelial cells and platelets stimulate vascular growth through the release of many angiogenic factors (e.g., VEGF, IL-8, FGF, PDGF), sustaining the nutritional needs of the tumor and favoring its survival and migration. (6) Numerous cytokines and factors released during inflammation can lead to metastatic events of tumors including IL-6 and IL-10. The inflammatory microenvironment has a major role in this setting with implications for the prevention and treatment of cancer. FGF, fibroblast growth factor; HIF, hypoxia-inducible factor; IL, interleukin; PDGF, platelet-derived growth factor; ROS, reactive oxygen species; VEGF, vascular endothelial growth factor; TNF-α, tumor necrosis factor. Parts of the figure were drawn by using pictures from Servier Medical Art. Servier Medical Art by Servier is licensed under a Creative Commons Attribution 3.0 Unported License (https://creativecommons.org/licenses/by/4.0/, accessed on 21 January 2024).
Cimb 46 00062 g001
Table 1. Detailed overview of the role of NGF and its receptors in various types of tumors. While NGF’s primary function is related to neural development and function, its relationship with tumors is complex and multifaceted. The involvement of NGF in tumors is not as straightforward as in normal nerve growth, and its effects on different types of tumors can vary. Acetylcholine, Ach; A disintegrin and metalloprotease 17, ADAM17; protein kinase B, Akt; extracellular signal-regulated kinase, ERK; F-box-only protein 22, FBOXO22; hypoxia-inducible factor 1 subunit alpha, HIF1α; nerve growth factor, NGF; non-small-cell lung cancer, NSCLC; neurotrophic tyrosine receptor kinase, NTRK; nuclear factor kB, NF-kB; p75 pan-neurotrophin receptor, p75NTR; programmed death-ligand 1, PD-L1; rearranged during transfection, RET; small nuclear ribonucleoprotein polypeptide A, SNRPA; tissue inhibitor of metalloproteinases, TIMP; tropomyosin receptor kinase, Trk; vascular endothelial growth factor, VEGF. (*) MYCN is amplified in 20% of neuroblastomas and correlates with aggressive phenotype and poor prognosis. (**) Perineural invasion driven by the TME has been identified as a key pattern of several malignancies including breast, pancreatic, and prostate cancers.
Table 1. Detailed overview of the role of NGF and its receptors in various types of tumors. While NGF’s primary function is related to neural development and function, its relationship with tumors is complex and multifaceted. The involvement of NGF in tumors is not as straightforward as in normal nerve growth, and its effects on different types of tumors can vary. Acetylcholine, Ach; A disintegrin and metalloprotease 17, ADAM17; protein kinase B, Akt; extracellular signal-regulated kinase, ERK; F-box-only protein 22, FBOXO22; hypoxia-inducible factor 1 subunit alpha, HIF1α; nerve growth factor, NGF; non-small-cell lung cancer, NSCLC; neurotrophic tyrosine receptor kinase, NTRK; nuclear factor kB, NF-kB; p75 pan-neurotrophin receptor, p75NTR; programmed death-ligand 1, PD-L1; rearranged during transfection, RET; small nuclear ribonucleoprotein polypeptide A, SNRPA; tissue inhibitor of metalloproteinases, TIMP; tropomyosin receptor kinase, Trk; vascular endothelial growth factor, VEGF. (*) MYCN is amplified in 20% of neuroblastomas and correlates with aggressive phenotype and poor prognosis. (**) Perineural invasion driven by the TME has been identified as a key pattern of several malignancies including breast, pancreatic, and prostate cancers.
Kind of TumorRole of NGFReferences
Brain tumors
  • Pro-NGF, NGF, and TrkA are expressed in numerous brain tumor cells (especially glioblastoma) and can promote cell survival and growth.
  • p75NTR proteolysis is required for brain tumor proliferation.
  • NGF concentrated in centrosome can phosphorylate TrkA, which may in return phosphorylate tubulin and promote mitotic spindle assembly, causing mitogenic effects in glioma cells.
  • In ependymoblastoma tumors, NGF exerts a marked action on differentiation rather than proliferation in vitro.
  • The potential of pro-NGF, NGF, and its receptors as clinical biomarkers and therapeutic targets has been highlighted.
[81,82,83,84,85,86,87,88,89]
Breast Cancer
  • NGF is both synthesized and released by breast cancer cells.
  • NGF exerts mitogenic, antiapoptotic, and angiogenic influences on breast cancer cells by engaging distinct signaling pathways that encompass the participation of TrkA and NGFR/p75NTR receptors.
  • Pro-NGF signaling has been linked to breast cancer invasion and metastasis.
  • NGF and its receptors have been identified as diagnostic and prognostic tools.
  • NGF and its receptors are promising therapeutic targets for breast cancer.
  • Norepinephrine/β2-Adrenergic Receptor pathway promotes cell proliferation and NGF production in triple-negative breast cancer.
  • Neurotoxin inhibition of sympathetic neural signaling in mammary tumors using 6-hydroxydopamine or genetic deletion of NGF or β2-adrenoceptor in triple-negative breast cancer tumor cells enhances the therapeutic effect of anthracycline chemotherapy by reducing metastasis in xenograft mouse models.
  • Electroacupuncture promotes apoptosis and inhibits axonogenesis by activating the p75NTR receptor for triple-negative breast xenograft in mice.
  • In rat models, it has been found that NGF from breast cancer may mediate spinal bone pain from metastasis via axonal growth and up-regulation of pain-associated neuropeptides.
[26,90,91,92,93,94,95,96,97,98,99]
Colorectal Cancer
  • NGF can stimulate the growth and survival of colon cancer cells and influence their invasive behavior.
  • NGF has been identified as a potential therapeutic target for the treatment of colon cancer.
  • NGF receptors may play a key role in androgen’s effect on hormone-sensitive tumor cells.
  • NGF has been included in new immunogenomic prognostic risk scores for colorectal cancer.
[17,47,100,101,102,103]
Gastric Cancer
  • In gastric cancer, NGF and Trk receptor mRNA expression are down-regulated.
  • NGF Trk receptors may elicit cell apoptosis by a Ras or Raf signal transduction pathway.
  • SNRPA (small nuclear ribonucleoprotein polypeptide A) enhances tumor cell growth in gastric cancer through modulating NGF expression.
  • ACh-NGF positive feedback loop may be the basis for the abnormal innervation observed in the TME and acts through the Trk receptors.
  • p75NTR inhibits the invasive and metastatic abilities of gastric cancer cells by down-regulating uPA and matrix metalloproteinase 9 proteins and up-regulating TIMP1 protein via the NF-kB signal transduction pathway.
  • p75NTR may be used as a new potential therapeutic target in metastatic gastric cancer.
  • Individual and co-expression patterns of NGF and heme oxygenase-1 may predict shorter survival of gastric carcinoma patients.
[104,105,106,107,108]
Head and Neck Cancer
  • NGF can stimulate morphological differentiation, adhesion, proliferation, and migration in head and neck cancer.
  • NGF controls cancer cell migration through Akt phosphorylation, suggesting a possible therapeutic role of Akt inhibitors.
  • NGF and TrkA are highly expressed in cases of head and neck carcinomas with and without perineural invasion and are associated with improved tumor cell survival. (**)
  • p75NTR and TrkC receptors demonstrate a different immunoreactivity profile in comparison to TrkA and TrkB receptors in the normal human pituitary gland and adenomas.
  • p75NTR and pattern of invasion predict poor prognosis in oral squamous cell carcinoma.
  • Pro-NGF may be a potential diagnostic biomarker for thyroid cancer.
  • In thyroid cancer, increased expression of the TrkA receptor has been correlated with tumor progression and lymph node invasion.
[10,44,109,110,111,112,113,114,115,116,117]
Leukemias
  • NGF induces the ERK signaling pathway through TrkA receptors stimulating the production and survival of immune cells, but the actual impact on hematological diseases is still to be determined.
[118,119,120,121]
Liver Cancer
  • NGF and TrkA are highly expressed in hepatocarcinoma tissue (especially in males).
  • p75NTR may provide a mechanism for selective apoptosis of hepatic stellate cells.
  • NGF and its receptors may play a role in cellular interactions involving hepatocarcinoma cells, hepatic stellate cells, arterial cells, and nerve cells in cancer tissues.
  • NGF regulates liver cancer cell polarity and motility associated with the invasion and metastasis process.
  • Various therapies for liver cancer have been found to act on NGF pathways or influence the pro-NGF/NGF balance.
  • NGF level evaluation may be useful to predict hepatic dysfunction after irradiation and has been studied as a possible biomarker for liver cancer.
[80,122,123,124,125,126,127,128,129,130,131,132,133]
Lung Cancer
  • NGF has been linked to lung cancer progression, promoting the growth and survival of lung cancer cells and contributing to the development of chemoresistance.
  • TrkA is increased in squamous cell carcinoma, NGF and pro-NGF are increased in both squamous cell carcinoma and in adenocarcinoma, and p75NTR is increased across all lung cancer histological subtypes compared to normal lung.
  • NGF might play a role in the interaction between lung cancer cells and nerve fibers, leading to increased tumor growth.
  • NGF and chemokines secreted by apoptotic astrocytes cause the formation of an inflammatory and immunosuppressive microenvironment, enabling the formation of a pre-metastatic niche in lung cancer brain metastases.
  • Evidence of Trk fusion in NSCLC suggests the potential of NGF receptors as targets for further therapeutical applications.
[83,134,135,136,137,138,139]
Ovarian Cancer
  • Ovarian cancer is marked by elevated levels of NGF and TrkA.
  • NGF is involved in perineural invasion. (**)
  • Through its interaction with the TrkA receptor, NGF decreases transcription of miR-145 levels, causing an increase in oncogenic proteins involved in promoting angiogenesis and cell proliferation and migration, as well as inhibiting apoptosis and influencing various molecules such as cyclooxygenase-2, ADAM17, and calreticulin, all essential for ovarian cancer progression.
  • MicroRNAs may be associated with NGF/TrkA activation and alter key protein levels.
  • The onset condition of ovarian cancer can be diagnosed through the detection of high or low expression of NGF and its receptors.
  • TrkA may be considered a new potential tumor marker.
  • NGF is also associated with increased resistance to chemotherapy in ovarian cancer cells.
  • Blocking neurotrophin action could be a therapeutic target in treating ovarian cancer.
  • Metformin treatment decreases the expression of c-MYC, β-catenin, and VEGF induced by NGF/TrkA while increasing oncosuppressor miRs, such as miR-145 and miR-23b.
[140,141,142,143,144,145,146,147,148,149]
Neuroblastoma
  • NGF promotes the survival and growth of neuroblastoma cells by binding to TrkA on the surface of cancer cells.
  • TrkA and TrkC are overexpressed in biologically favorable neuroblastomas: their expression was associated with an absence of N-myc amplification, lower disease stage, lower patient age, differentiated tumors, and a greater likelihood of spontaneous regression or responding well to therapy.
  • TrkB is mainly expressed in unfavorable, aggressive neuroblastomas.
  • p75 plays an important role in enhancing both the sensitivity of Trk receptors to low levels of ligand, as well as enhancing ligand-induced differentiation in TrkA/p75 but not TrkB/p75 cells.
  • Targeting Trk receptors is a potential therapeutic strategy for neuroblastoma treatment.
  • MYCN (*) targets estrogen receptor alpha (Erα) and thereby NGF signaling to maintain an undifferentiated and aggressive phenotype.
  • RET and TrkA physically interact and can induce reciprocal activation in response to ligand activation.
[24,150,151,152,153,154,155,156]
Pancreatic Cancer
  • NGF exerts both stimulatory and inhibitory effects on pancreatic cancers with the effect based on the expression levels and the ratio of TrkA and p75NTR.
  • NGF from pancreatic stellate cells induces pancreatic cancer proliferation and invasion by the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/glycogen synthase kinase (GSK) signal pathway.
  • TrkA expression in pancreatic cancer is a marker of tumor aggressiveness.
  • Elevated p75NGFR expression is associated with a favorable prognosis.
  • NGF overexpression combined with TrkA may contribute to perineural invasion by activating the Warburg effect, promoting tumor-derived exosomal miRNA-21 expression, prompting the hyperplasia of nerves, and inhibiting tumor cell apoptosis. (**)
  • In pancreatic cancer, the high-glucose microenvironment promotes the invasion ability and raises the expression of NGF by upregulating HIF1α.
  • NGF has been successfully used for diagnostic, therapeutic, and prognostic purposes in pancreatic cancer.
  • Atorvastatin may exert an anti-tumor effect in pancreatic cells via the inhibition of NGF and other neurotrophin signaling pathways.
  • In a mouse model, anti-NGF treatment beginning at 4 weeks may increase inflammation and negatively impact disease, while treatment starting at 8 weeks (after disease onset) reduces neural inflammation, neural invasion, and metastasis.
[13,15,16,157,158,159,160,161,162,163,164,165,166,167]
Pediatric tumors
  • NGF Trk receptors play a key role in pediatric tumors, especially in brain cancers.
  • Trk receptors play a key role in transducing the mitogenic effects of NGF and Trk inhibitors have been proven to be an important therapeutic tool for the treatment of NTRK fusion cancers.
  • NGF administration may be an effective and safe adjunct therapy in children with optic atrophy due to optic gliomas.
[28,36,82,168,169,170]
Prostate Cancer
  • NGF and its receptors are found in prostate cancer cells.
  • NGF is released by both epithelial pancreatic cells and cancer-associated fibroblasts.
  • NGF may be able to suppress tumor growth via an indirect effect, probably innervation or maturation of the tumor neo-vasculature.
  • Aberrations and/or derangement of NGF signaling contribute to tumor growth and progression, enhancing the invasive potential of prostate cancer cells and promoting the development of cancer cells’ neuroendocrine features.
  • Expression of p75NTR reduces NGF-induced cell growth by activation of programmed cell death.
  • Cross-talk between androgen receptors and NGF receptors in prostate cancer cells may have implications for a new therapeutic approach.
  • Pro-NGF correlates with the Gleason score and is a potential driver of nerve infiltration in prostate cancer.
  • Cancer-associated fibroblasts can activate Yes-associated protein (YAP1)/TEA domain (TEAD1) signaling and increase the secretion of NGF, therefore promoting perineural invasion. (**)
  • FBXO22 mediates the NGF/TrkA signaling pathway and stimulates macrophage M2 polarization in prostate cancer bone metastases, promoting cell activity and osteogenic lesions.
[77,171,172,173,174,175,176]
Skin tumors
  • NGF and its receptor seem to play a role in most skin tumors including malignant melanoma.
  • In basal cell carcinoma and cutaneous squamous cell carcinoma, increased levels of NGF and TrkA, B, and C may reflect unique survival pathways.
  • P75NTR may play a mechanistic role in invasive melanomas demonstrating perineural invasion. (**)
  • Higher levels of Trk receptors in cutaneous squamous cell carcinoma cells may predict perineural invasion.
  • Increased p75NTR expression in cutaneous squamous cell carcinoma perineurally may allow p75NTR immunohistochemical staining to be used for detecting sites of perineural invasion.
  • NGF and TrkA are overexpressed in cervical squamous cell carcinoma.
  • PD-L1 and NGF are co-expressed on spindle cells in the microenvironment of Merkel carcinoma, and TrkA receptors seem to play a major role.
  • P75NTR has been identified as a useful marker to distinguish spindle cell melanoma from other spindle cell neoplasms of sun-damaged skin.
[54,177,178,179,180,181,182,183,184,185,186]
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

Ferraguti, G.; Terracina, S.; Tarani, L.; Fanfarillo, F.; Allushi, S.; Caronti, B.; Tirassa, P.; Polimeni, A.; Lucarelli, M.; Cavalcanti, L.; et al. Nerve Growth Factor and the Role of Inflammation in Tumor Development. Curr. Issues Mol. Biol. 2024, 46, 965-989. https://doi.org/10.3390/cimb46020062

AMA Style

Ferraguti G, Terracina S, Tarani L, Fanfarillo F, Allushi S, Caronti B, Tirassa P, Polimeni A, Lucarelli M, Cavalcanti L, et al. Nerve Growth Factor and the Role of Inflammation in Tumor Development. Current Issues in Molecular Biology. 2024; 46(2):965-989. https://doi.org/10.3390/cimb46020062

Chicago/Turabian Style

Ferraguti, Giampiero, Sergio Terracina, Luigi Tarani, Francesca Fanfarillo, Sara Allushi, Brunella Caronti, Paola Tirassa, Antonella Polimeni, Marco Lucarelli, Luca Cavalcanti, and et al. 2024. "Nerve Growth Factor and the Role of Inflammation in Tumor Development" Current Issues in Molecular Biology 46, no. 2: 965-989. https://doi.org/10.3390/cimb46020062

Article Metrics

Back to TopTop