Next Article in Journal
Vulvar Paget’s Disease: A Systematic Review of the MITO Rare Cancer Group
Previous Article in Journal
Inhibition of IDH3α Enhanced the Efficacy of Chemoimmunotherapy by Regulating Acidic Tumor Microenvironments
Previous Article in Special Issue
HIPK2 in Angiogenesis: A Promising Biomarker in Cancer Progression and in Angiogenic Diseases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Genomic Interplay between Neoneurogenesis and Neoangiogenesis in Carcinogenesis: Therapeutic Interventions

1
SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Pretoria 0028, South Africa
2
Department of Medical Oncology, Faculty of Health Sciences, Steve Biko Academic Hospital, University of Pretoria, Pretoria 0028, South Africa
3
Department of Surgery, Steve Biko Academic Hospital, University of Pretoria, Pretoria 0028, South Africa
4
Centre for Cancer Sciences, Division of Cancer and Stem Cells, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
*
Authors to whom correspondence should be addressed.
Submission received: 1 February 2023 / Revised: 8 March 2023 / Accepted: 13 March 2023 / Published: 16 March 2023
(This article belongs to the Special Issue New Advances in Tumor Metastasis Angiogenesis-Dependent Processes)

Abstract

:

Simple Summary

This review describes how the two processes of angiogenesis (the generation of new blood vessels) and neurogenesis (the generation of new nerve fibers) act together to drive cancer progression. It also describes how they are both associated with a lower rate of patient survival. These two processes share signaling pathways and, in many cases, the initiation of one leads to the initiation of the other. Both processes require tissue alterations and are reliant on cell migration. They favor cancer progression by supplying the tumor with nutrients and facilitating communication/movements within the tumor. Thus, these two processes contribute to the spread of cancers, as tumors can use nerve fibers and blood vessels as a routes to migrate from the initial point of cancer development to the surrounding area or to the distal sites of the body.

Abstract

Angiogenesis, the generation of new blood vessels, is one of the hallmarks of cancer. The growing tumor requires nutrients and oxygen. Recent evidence has shown that tumors release signals to attract new nerve fibers and stimulate the growth of new nerve fibers. Neurogenesis, neural extension, and axonogenesis assist in the migration of cancer cells. Cancer cells can use both blood vessels and nerve fibers as routes for cells to move along. In this way, neurogenesis and angiogenesis both contribute to cancer metastasis. As a result, tumor-induced neurogenesis joins angiogenesis and immunosuppression as aberrant processes that are exacerbated within the tumor microenvironment. The relationship between these processes contributes to cancer development and progression. The interplay between these systems is brought about by cytokines, neurotransmitters, and neuromodulators, which activate signaling pathways that are common to angiogenesis and the nervous tissue. These include the AKT signaling pathways, the MAPK pathway, and the Ras signaling pathway. These processes also both require the remodeling of tissues. The interplay of these processes in cancer provides the opportunity to develop novel therapies that can be used to target these processes.

1. Introduction

The hallmarks of cancer are characteristic alterations in the cells that accompany or lead to the development and progression of cancer. One of these hallmarks is uncontrolled or dysregulated angiogenesis. This allows the tumor mass to be provided with nutrients as a result of the development of new blood vessels (reviewed in [1]). Recently, evidence brought forward shows how the creation of new nerve tissue, neurogenesis, is another important contributing factor to the development and progression of cancer [2,3,4]. Multiple studies indicate that tumors in tissue with a high level of blood vessels present are more likely to develop intra-tumoral neural infiltration, a condition that is associated with a poor prognosis. The more extensive this intra-tumoral nerve density is, the more severe the metastatic potential of the tumor and the poorer the prognosis is for the patient [5,6,7]. In the body, the distribution of blood vessels and nerves mirrors each other (Figure 1A). This is due their embryological and growth factor similarity - they share many of the same molecules and signaling pathways that guide their growth [8]. These shared molecules and pathways may be the reason why the molecules that induce angiogenesis during cancer also induce neurogenesis. Examples of the interaction between the nervous tissue and the surrounding blood vessels include the blood brain barrier (BBB), where astrocytes and pericytes around blood vessels interact with these blood vessels to aid in the regulation of the BBR. Interactions between the nervous tissue and the blood vessels surrounding it are known as neurovascular units (NVUs) (Figure 1B) [9]. Brain tumors are known to alter the structure of the blood brain barrier (BBB) into what is known as the blood–tumor barrier (BTB). It is now known that cancers can drive neurogenesis, axonogenesis, and the repurposing of existing nerve fibers.
The new nervous tissue formed during neurogenesis is also dependent on new blood vessels to supply the nervous tissue with oxygen and nutrients to ensure the new tissues’ continued survival [10]. This review discusses the role played by the interplay between neurogenesis and angiogenesis in cancer, and although many of the examples discussed come from studies on the interplay of these processes following stroke, the pathways discussed are expected to be the same or similar to the pathways exploited by cancer cells to attract or create nerve fibers or increase the vascularization to allow tumors to grow and metastasize. During metastasis, cancer cells access the circulatory system, move, and implant in distant organs. This process involves reciprocal communication between cancer cells, endothelial cells of the vasculature, and neural cells of the nervous systems. This communication correlates with poor prognosis [11].

2. Angiogenesis and Neurogenesis

The reciprocal communication between tumors and the nervous system is evidenced by the fact that cancer patients experience cancer related pain [12] as a result of neuro-oncogenic pressure on the fibers as the tumor volume increases [13], secretion of stimulatory factors on peripheral fibers with depolarizing effects, axon demyelination, and pathological neural plasticity induced by tumor-derived factors [14].
The term angiogenesis describes the generation of new blood vessels. There are two main methods whereby angiogenesis occurs. Vasculogenic involves vascular progenitor cells forming new blood vessels. These circulating endothelial progenitor cells (EPCs) are derived from the bone marrow [15]. These cells possess surface markers, such as CD34, CD31, VEGFR-2, and Tie-2. Tumor cells are known to secrete VEGF and cytokines such as stromal-derived factor-1. These pro-angiogenic factors recruit the circulating progenitor cells and stimulate proliferation. These pro-angiogenic stimuli also activate the matrix metalloproteases to break down the extracellular matrix, allowing cells to migrate and penetrate cell layers [16].
Sprouting angiogenesis involves the formation of new vascular structures from an existing vessel. This, firstly, involves the destabilization of endothelial cells and endothelial mesenchymal transition. Once again, activated proteases are required to degrade the ECM and basement membrane, allowing for directed migration and proliferation [16]. Once the cells form the lumen and tubulogenesis of the new blood vessels, the transition is reversed, with the cells reverting to the resting state [17]. This process involves the VEGF and Notch signaling pathways [18]. As such, sprouting angiogenesis is a process whereby new blood vessels are formed from pre-existing blood vessels. This process occurs in response to both mechanical and chemical stimuli and is important for normal development and wound healing. It facilitates tumor survival and progression as it provides cancer cells with oxygen and nutrients to sustain their growth (reviewed in [19]). During angiogenesis, new micro vessels are formed as they branch off from pre-existing vessels [20].
Angiogenesis involves processes, such as proliferation of endothelial cells and the formation of tube-like vascular structures, as well as branching and anastomosis (reviewed in [21]). This review will mainly focus on sprouting angiogenesis. Both pro- and anti-angiogenic factors act to regulate angiogenesis, with any changes in the balance of these factors either promoting or inhibiting angiogenesis. Pro-angiogenic factors are released by endothelial cells, monocytes, and tumor cells. Angiogenesis also requires the activation of various processes, such as basement membrane degradation, endothelial cell proliferation, migration, and cell remodeling, to form a tube or blood vessel (reviewed in [22]). Angiogenesis can be promoted by adrenergic signaling, which is thought to contribute to the formation of preneoplastic niches and decreased immune function [23]. β-adrenergic receptor activation has been found to be essential in the progression and malignant growth of ovarian [24], pancreatic [25], and pulmonary [26] cancers.
In adult mice, neurogenesis predominantly occurs in the subventricular zone (SVZ) and the hippocampal dentate gyrus (DG), and it is thought that these two areas are where neural stem cells are found within the adult brain [27,28]. Different types of cells are involved in neurogenesis. The slow-dividing or true stem cells are mostly quiescent but activate and divide asymmetrically to self-renew and give rise to immediate progenitor cells [27]. These intermediate progenitor cells divide rapidly to give rise to nervous tissue. In addition to these progenitor cells, other types of cells integrate into existing neuronal networks. These include neuroblasts [29,30] and newborn neurons, which are electro-physiologically active [31]. Neurogenesis can be activated or inhibited by a multitude of signals, including growth factors, cytokines, chemokines, neurotrophins, steroids and extracellular matrix components, the activation of specific transcription factors, and signal transduction pathways (reviewed by [32]). Additional signals may result from environmental stimuli, such as exercise alterations in an organism’s environment, stress, or social isolation [33,34]. The growth and migration of tumors around and along nerve fibers are known as perineural invasion [35]. Additionally, it is now known that nerves actively grow into and throughout cancer tissue, resulting in increased metastasis as these nerves provide signals to the cancer as well as a pathway to migrate along [36]. Autonomic neurotransmitter receptors can stimulate cancer cell growth through the activation of corresponding signaling pathways [37]. In its most basic form, the crosstalk between neurogenesis, angiogenesis, and carcinogenesis is based upon common signaling pathways and chemokines. Cancer cells express neurotrophic and pro-angiogenic markers. In many cases, these molecules are both neurotrophic and pro-angiogenic. The infiltration of new nervous tissue into cancer cells gives cancer cells a route to migrate along in the same way that these cells can use lymphatic and blood vessels. In other words, the signaling pathways activated by more cells stimulate both neurogenesis and angiogenesis while assisting in metastasis [38].

3. Stem Cell Niches and Blood Vessels

In adults, the stem cells that give rise to new tissue are in microenvironments known as stem cell niches. It is the communication between the stem cells and these niches which governs the activity of these stem cells, instructing them when to proliferate and differentiate. This also involves niche cells regulating stem cell activity in response to external signals.
This close association between blood vessels and stem cells has been observed for hematopoietic stem cells [39] and neuronal stem cells [40,41,42]. When it comes to neurogenesis, neural stem cells, otherwise known as neural progenitor cells, migrate through blood vessels to the tumor site from the neurogenic regions of the brain and metastatic niches. These cells express the doublecortin (DCX) surface marker for progenitor cells, which has been found to be expressed at higher levels in individuals with high-risk cancers. Once these cells reach the tumor site, they differentiate into various noradrenergic, mature neuronal cells [43]. Cancer stem cells can also form new neurons that are themselves capable of stimulating tumor growth in xenograft models [44]. These neural stem cells are surrounded by an extracellular matrix, rich in both blood vessels and laminin. Neurogenesis can also be induced or inhibited through direct physical contact and, therefore, signaling via integrins between the blood vessels and neural stem cells. One of these signals is the laminin receptor for α6β1integrin, which is expressed by both the stem cells and blood vessels [41,45]. Following the inhibition of α6β1integrin, the levels of both stem cell proliferation and migration away from blood vessels increase [41]. Blood vessels nearby and towards the SVZ vascular niche can interact with the SVZ, leading to the release of angiogenic factors from the surrounding endothelial cells, resulting in neurogenesis [41]. One of these factors is the chemokine CXCL12 (SDF1), which initiates a signaling pathway, resulting in the migration of neural stem cells to blood vessels and surrounding endothelial cells that express CXCL12 [46]. Endothelial cells are also able to secrete factors that inhibit neurogenesis. These include sphingosine-1-phosphate and prostaglandin-D2, both of which are G-protein-coupled receptors (GPCRs), ligands that keep neural stem cells in a quiescent state [47]. Another factor secreted from endothelial cells is neurotrophin-3 (NT-3), which is required to sustain and regulate neurogenesis [48].

4. Ischemic Stroke Models Providing Evidence for the Interaction of Neurogenesis and Angiogenesis

The first evidence showing the association between neurogenesis and angiogenesis was from stroke models. In these models, it was noted that recovery from the stroke resulted in increased numbers of neural stem cells and increased levels of angiogenesis [49,50]. Many of these models demonstrated the influence of therapies to treat stroke victims on the processes of neurogenesis and angiogenesis, for example, the use of stem cell therapy. Rat models of stroke showed that mesenchymal stem cell (MSC) therapy is due to factors secreted by these stem cells rather than these cells replacing lost tissue. The effect of these secreted factors rather than the cells was tested using a Wistar rat model of stroke where the rats underwent cerebral artery occlusion. These secreted factors were able to increase the levels of neurogenesis and angiogenesis in these rats, which was associated with a decrease in the neurodegenerative effects following stroke [51]. Surgical interventions for the treatment of stroke, namely, deep brain stimulation (DBS), which involves the electrical stimulation of nerve tissue, were performed. Rat models of ischemic stroke showed that recovery from stroke following DBS treatment was associated with increased proliferation and movement of cells from the subventricular zone. This is accompanied by an increase in the expression of growth factors and the stimulation of endogenous neurogenesis and angiogenesis [52]. The migration of neural progenitor cells has been shown in these models to be closely associated with blood vessels, with these vessels guiding the movement of these cells [53]. The expression of genes, miRNAs, and lncRNAs associated with these processes and recovery following ischemic stroke has shown that there are many complex signaling processes involved in neurogenesis, angiogenesis, and final recovery (reviewed in [10]).
The use of these rat models identified many important molecules involved in angiogenesis, neurogenesis, and cell movement. These include miRNA-9, which links neurogenesis and angiogenesis through regulating expression of VEGF-A by targeting transcription factors that induce VEGF-A expression [54]. Mouse models demonstrated that miRNA-126 promotes proliferation, migration, angiogenesis, and neurogenesis after brain ischemia by inhibiting its target tyrosine–protein phosphatase non-receptor type 9 (PTPN9) and activating AKT and ERK signaling pathways [55]. Signaling pathways that were identified to play a role in these processes include STAT3 pathways [56], sonic hedgehog [57], and EphA receptor-mediated signaling [58].

5. The Remodeling of Tissues and the Proliferation and Migration of Cells during Angiogenesis and Neurogenesis

When malignant cells invade surrounding tissues, they displace the normal cells or integrate them, altering their function. These cells that are co-opted by cancer cells to contribute to the survival of the tumor include fibroblasts [59], endothelial cells [60], immune cells [61,62], and neuronal extensions [63]. Metastatic tumors then need to form capillaries to supply oxygen and nutrients but also to act as routes for further cell movement for metastatic dissemination [64]. Nerve fibers can provide similar functions by providing the cancer cells with nerve cell signals as well as serving as migrations routes [65]. Both nerve cells and the endothelial cells that become blood vessels are attracted to and co-opted by the cancer cells [66]. Neurotrophins, such as nerve growth factor (NGF), can be released by leukocytes, such as macrophages and mast cells, to promote an axonogenic switch resulting in tumor innervation. Many immune cells express NGF [67] following induction by IL-1β [68]. This can occur during inflammatory pain and neurogenesis. In a mouse model of arthritis, the activation of macrophages results in increased levels of innervation, related to and in conjunction with angiogenesis [69].
Neurogenesis and angiogenesis require increased proliferation of neural and endothelial cells. This can be achieved by altering the length of the cell cycle, shortening it, and allowing for more cycles of division [70]. Alternatively, the rate of different types of divisions can be altered since symmetric divisions will increase the number of stem cells, while asymmetric division will result in a differentiated cell and one stem cell being generated [71].
The beta-adrenergic receptors, β2/3 receptors, have been shown to be involved in tumor development and progression. In a mice model, the lack of the β2 or β3 receptor led to a delay in tumor growth and angiogenesis [5,72]. In prostate cancer, these receptors lead to the stimulation of endothelial cells, resulting in angiogenesis by metabolic adjustments. The sympathetic nerves release noradrenaline, which activates β2-signaling in endothelial cells, leading to the expression of the mitochondrial cytochrome c oxidase component, COA6. This results in a decrease in normal oxidative respiration and the induction of angiogenesis [72]. This noradrenaline also promotes vascular endothelial growth factor (VEGF) expression, leading to angiogenesis [73].

5.1. Migration and Remodeling

Both angiogenesis and neurogenesis require the initiation or alteration of cell migration as well as the remodeling of tissue, which, themselves, require the breakdown of the extracellular membrane. In order to accomplish this altered migration and tissue remodeling, there must be specific interactions between cells, such as the immature migrating neuroblasts, astrocytic processes, and blood vessels [74]. Some of the molecules involved in these processes include stromal-cell-derived factor-1 (SDF-1), CXC chemokine receptor 4 (CXCR4), monocyte chemoattractant protein-1 (MCP-1), and matrix metalloproteinases (MMPs).

5.2. SDF-1 and CXCR4

The alpha chemokine SDF-1 protein is normally excreted from the ependymal cells; it is also known as CXCL12 and, under normal conditions, induces neural stem cell (NSC) quiescence [75]. However, under abnormal conditions such as in ischemic shock, SDF-1 is released from reactive astrocytes and vascular cells and, in these cases, leads to increased activation of NSCs [46]. The receptor for SDF-1, CXCR4, is also normally expressed in bone marrow, where both CXCR4 and SDF-1 are involved in hematopoietic stem cell mobilization and trafficking of NSCs [76]. In a similar way, it is thought to mobilize and direct neuroblasts. The migration of NSCs in a rat stroke model was inhibited following the blocking of SDF-1 function using an antibody against CXCR4 [77]. SDF-1 may be one of the factors released by endothelial cells that leads to the directing of migrating neuroblasts to the vasculature [46].

5.3. MCP-1

The (CC) family member chemokine, monocyte chemoattractant protein-1 (MCP-1), is known to interact with the CC-chemokine receptor-2 (CCR2). CCR2 is widely expressed on NSCs, and the binding of MCP-1 leads to increased NSC migration in vitro [78]. It is suspected that this is accomplished through the receptor and ligand activating the PI3 kinase pathway [79].

5.4. MMPs

Matrix metalloproteinases (MMPs) are known to have a role in cancer, promoting metastasis through the breakdown of the extracellular matrix (ECM). This then allows cells to migrate through tissue layers. This family of proteases also seems to play a role in the migration of SVZ neuroblasts. MMPs-3 and -9 are expressed in neuroblasts and their inhibition results in reduced neuroblast migration [80]. MMPs-2 and -9 act on the vasculature to assist in the migration of neuroblasts by activating the PI3K/Akt and ERK1/2 signaling pathways [77].

6. Growth Factors

Growth factors are responsible for guiding the growth of both blood vessels and nerve fibers to new target tissues or destinations. These growth factors include VEGF, NGF, and insulin growth factor (IGF). Fibroblast growth factor 2 (FGF2) stimulates neurogenesis [81], whilst bone morphogenic protein 4 (BMP4) has an inhibitory effect on neurogenesis [82]. The pituitary-gland-derived hormone, prolactin, induces neurogenesis [83], while growth differentiating factor 1 (GDF11) augments neurogenesis in older adults where the normal levels of neurogenesis have declined [28]. Brain-derived neural factor (BDNF) and pigmented epithelium-derived factor (PEDF) are both secreted from the blood vessels and endothelial tissue and both induce neurogenesis [84]. Tumor-associated neoneurogenesis involves the activity of growth factors, which generally have pleiotropic functions and serve to attract nerve fibers to a tumor and initiate neural cell growth [7]. Cancer cells also release axon guidance molecules (netrins) to aid in nerve infiltration [85]. Neurotrophic growth factor receptors and neurotransmitter receptors (NTRs) are both expressed in neurons and cancer cells. This gives their ligands, growth factors, and neurotransmitters the ability to act as signals that connect nerves and cancer cells. Axon guidance or the generation of new axons into a tumor and angiogenesis can both be guided by neurotrophins. This was demonstrated using the expression patterns of the Tyrosine receptor kinases (Trks), with different Trk receptors binding different neurotrophins: NGF binds to TrkA, BDNF binds to TrkB, and neurotrophin 3 (NT3) binds to TrkC. The expression of these Trks was established in tumors from oral squamous cell carcinoma (OSCC) patients as well as on the surface of high metastatic cells grown in culture. In both cases, the Trk receptors TrkB and TrkC were overexpressed. The expression levels of these receptors in OSCC patients correlated with a poor prognosis and lower survival rates [86]. The same was true for increased expression of TrkB in patients with ovarian cancer [87]. Some neurotrophies, such as NGF and BDNF, can promote angiogenesis independently of VEGF and may be the reason why some tumors are resistant to anti-VEGF therapy [88].
Neurotransmitters can also link neurogenesis, angiogenesis, and cancer. Increased levels of the dopamine receptor D2R are associated with lower survival rates in gastric cancer [89]. Dopamine is also able to stimulate the EGFR-AKT pathway, stimulating migration and invasion [24]. The increased expression of the D2R receptor has been observed in multiple cancers [90,91].

6.1. FGF2

Fibroblast growth factor (FGF-2) and its receptor FGFR2 are able to stimulate endothelial cell proliferation [92]. FGF2 is known to be overexpressed in dividing neural stem cells and its overexpression leads to an increased proliferation of neural stem cells [93]. It has also been shown that increased FGF-2 expression in a rat stroke model leads to increased neurogenesis and can lead to the recovery of brain function following a stroke [94]. Upregulation of IGF-1 is also observed in angiogenesis [92]. It is also responsible for regulating migration and ECM degradation during angiogenesis where it increases the expression of Urinary-Type Plasminogen Activator and matrix metalloproteinases in endothelial cells [95]. FGF2 stimulation of endothelial cells leads to the formation of membrane vesicles containing MMP-2, MMP-9, TIMP-1, and TIMP-2 on the cell surface. These vesicles assist in the formation of capillary-like structures [96]. Like many other growth factors, the activation of the Ras/ERK pathway by the binding of FGF3 to FGFR results in the expression of pro-angiogenic and pro-neurogenic genes (Figure 2).

6.2. IGF-1

Insulin-like growth factor-1 (IGF-1) is known to play a role in the development of the brain; despite this, it is normally expressed in the liver. IGF-1 is able to induce the proliferation of adult neural stem cells in the hippocampus. This proliferation occurs as a result of the activation of the MAPK [97]. Not only does IGF-1 have neurogenic/neurotrophic properties but it is also able to induce angiogenesis [98]. An experiment using a mouse model of permanent focal ischemia was used to illustrate the overexpression of IGF-1 using an adeno-associated viral (AAV) vector with the IGF-1 gene cloned into the vector. This overexpression resulted in higher levels of vascular density and increased neurogenesis noted the day after ischemic injury. This is accomplished through IGH-1 activating ERK, which leads to the generation of vascular endothelial cells and the initiation of PI3K and AKT signaling (Figure 3) [99]. IGF-1 also induces the activity of the Brain-4 (Brn4) transcription factor. This leads to the promotion of neural differentiation in neuronal stem cells in the hippocampus [49].

6.3. VEGF

Vascular endothelial growth factor induces angiogenesis after binding to its receptor on endothelial cells. The role played by VEGF in angiogenesis is well-understood but recent evidence suggests that it also plays a role in neuronal growth, survival (neurotrophic), axonal outgrowth (neurotropic), and neuroprotection. It has been established that VEGF has direct effects on neurons and glial cells, leading to increased growth (including axonal outgrowth) and survival (Figure 4). VEGF has also been implicated in multiple neurological disorders [100]. These direct effects are due to the presence of VEGF receptors on the surface of neuronal cells. These receptors include Flk-1 and Flt-1 [101].

6.4. BDNF

Brain-derived neurotrophic growth factor is known to induce neurogenesis. It has been shown that intrahippocampal administration of BDNF increased neurogenesis in an adult rat model. Intracerebroventricular infusion of BDNF led to increased production of new nerve cells in the olfactory bulb of adult rats [102]. Overexpression of BDNF using an AAV vector resulted in higher levels of neurogenesis, and in a mouse model of stroke injury resulted in better recovery following ischemic injury [103]. In mouse models of myocardial infarction, some mice were wild-type while others were BDNF (+/−) heterozygous. The heterozygous mice showed better recovery rates as well as reduced levels of vascularization. This indicates that BDNF is able to induce angiogenesis [104] (Figure 5). BDNF stimulates angiogenesis by recruiting bone-marrow-derived cells as endothelial progenitor cells. BDNF also stimulates stem cells to differentiate into endothelial cells [105].

6.5. NGF

The attraction of nerve fibers to tumor cells is facilitated by cancer cells secreting neurotrophic growth factors. [roNGF is expressed in prostate cancer cells, where, once it is processed to nerve growth factor, drives nerve infiltration. The levels of proNGF correlated with tumor aggressiveness, with low-risk tumors showing significantly lower levels [106]. Prostate cancer cells are also able to stimulate neuron outgrowth by releasing proNGF (Figure 6) [106]. There are some doubts as to whether proNGF needs to be processed into NGF to perform these functions, with some researchers believing that proNGF itself is responsible for these actions. Other processes regulated or stimulated by proNGF/NGFs include perineural invasion [7] and tumor neoneurogenesis [106].
The role played by NGF as a neurotrophin, promoting neurotrophic and neurotropic effects in sympathetic neurons, is well known. Recent evidence suggests that NGF also plays a role in angiogenesis. Angiogenesis involves changes and migration in endothelial cells (ECs). It was found that NGF was able to induce the migration of cultured endothelial cells to the same extent as VEGF [107]. Another study reported similar results using the chorioallantoic membrane (CAM) of a quail as a model system. Due to the highly vascularized nature of this membrane, quail is an ideal model to study angiogenesis. NGF was found to have pro-angiogenic effects on the natural vascularization of these membranes and, once again, this effect was similar to that induced by recombinant VEGF165a (rhVEGF) [108]. These effects of NGF on EC migration and CAM vascularization could be blocked by the addition of the NGF receptor antagonist K252a[(8R*,9S*,11S*)-(/)-9-hydroxy-9-methoxycarbonyl-8-methyl-2,3,9,10-tetrahydro-8,11-epoxy-1H,-8H,11H-2,7b,11a-triazadibenzo(a,g)cycloocta(c,d,e)trindene-1-one]. This selective blocker targets the NGF/trkA receptor [107,108]. The similar VEGF blocker SU-5416, which targets the VEGF/Flk1 receptor, had no effect on the proangiogenic effect of NGF. Therefore, NGF has a direct angiogenic effect, relying on a distinct signaling pathway from the pro-angiogenic effect induced by VEGF (Figure 6) [108]. However, the study on the effects of NGF on EC migration indicated that the signaling pathways activated by the growth factors are able to activate each other’s downstream tyrosine kinase signaling pathways [107]. NGF also promotes VEGF expression through the MAPK-ERK2 phosphorylation signaling pathway [109].

6.6. Nestin

Nestin is a class VI intermediate filament that is normally expressed in nervous tissue in mammalian embryos. It is also found in the adult brain where it is found in the subventricular zone. This zone is where neurogenesis occurs. Nestin plays a role in brain development where it promotes survival, renewal, and mitogen-stimulated proliferation of neural progenitor cells. It also plays a role in mitosis, where it disassembles phosphorylated vimentin intermediate filaments (IFs). Nestin expression is also upregulated in various types of tumors. In these tumors, nestin expression was found to occur in the vascular endothelial cells, the same areas where angiogenesis takes place [110].

6.7. Neuropilins

The neuropilins (NRPs) are plasma membrane spanning receptors that have no cytosolic protein kinase domain, meaning that they only function as co-receptors of other receptors for various ligands. They were originally identified as co-receptors for semaphorin (reviewed in [111]) and vascular endothelial growth factor [112]. By acting as a co-receptor for semaphorins, they play a role in axon guidance during axon and neurogenesis, while as co-receptors for VEGF, they play a role in angiogenesis. Commonly, this role in angiogenesis is related to providing new neurons with blood supply [113,114]. In addition, they also contribute to the regulation of signaling pathways, such as the STAT, RAS, MAPK, PI3K, Notch, TGF-β, Wnt/β-catenin, and hedgehog pathways. This allows them to play a role in processes, such as remyelination, immune response, angiogenesis, cell survival, migration, and invasion [115,116,117]. The expression of NRP is known to be altered in tumors [118], with more advanced-stage tumors showing higher levels of expression [119,120]. The overexpression of these NRPs leads to leaky hypervascularization [121].
There are two members of the family, Neuropilin-1 (NRP-1) and Neuropilin-2 (NRP-2) [122]. NRP1 is known to be vital for the vascularization of the spinal cord, hindbrain, and retina in mouse models [121,123,124]. This confirmed the need for NRP1 in the vascularization of the CNS. This is due to NRPs acting as an adhesion molecule and receptor for two particular classes of semaphoring and VEGF, namely class 3 semaphorin (SEMA3A) and VEGF165a [125]. Alternative splicing gives rise to both membrane-bound and soluble isoforms of NRPs. Different forms of NRP are also created by the inclusion of different lengths of amino acids to the C-terminal [126].

7. The Role of Reactive Oxygen Species

As they multiply, malignant cells require nutrients and oxygen to survive and continue growing (reviewed in [127]). Cancer cells that are deprived of oxygen respond to local hypoxic conditions by releasing signaling molecules, such as chemokines and growth factors, that alter the tumor microenvironment. These signaling molecules interact with immune, endothelial, and neuronal cells and induce their migration to the primary tumor [60,128]. Angiogenesis is also regulated via the crosstalk between reactive oxygen species (ROS) and Ca2+ ions. The ROS, nitric oxide, activates Ca2+ channels. This occurs via the glutathionylation of the Serine incorporator (SERINC) protein and the involvement of the NADPH oxidase (NOX) protein family. This process is also known as the VEGF-dependent anti-ROS angiogenic pathway as VEGF can induce this process. This is demonstrated by the induction of endothelial cell migration by H2O2, which, in turn, can be inhibited by catalase and superoxide dismutase (SOD). The migration of these endothelial cells is also accompanied by an influx of Ca2+ into these cells [129]. This process is reliant on NOX2 or Nox4 activity. H2O2 supplementation can bypass the lack of NOX4 but not Nox2. This indicates that NOX2 is acting downstream of both H2O2 and Nox4. The production of ROS by Nox4, in turn, is activated by VEGF. Certain ROS can also activate the expression or stimulate the activity of transcription factors that regulate angiogenesis [130].
The expression of both NOX2 [131] and Nox 4 [132] has been found to be expressed in neurons. Nox1 expression was also found to increase in response to nerve growth factor (NGF) signaling [133]. In neurons, NADPH oxidases play roles in the modulation of the activity of neurons as well as altering the cell fate or neurons. Brain-derived neurotrophic factor signals for neurons to undergo apoptosis as a result of serum deprivation, a process which is modulated by NOX2 [134]. NOX1 also negatively regulates neurite outgrowth [133].
Ets-1, NF-kB, and STAT-3 ROS also induces the expression of various genes involved in angiogenesis, such as monocyte chemoattractant protein-1 (MCP-1), vascular cell adhesion molecule 1 (VCAM-1), and matrix metalloproteinases (MMPs).
The contribution of the nervous system to cancer progression was demonstrated in mouse models of gastric cancer. Here, the surgical or pharmacological denervation of the stomach suppressed gastric tumorigenesis [135]. Denervation resulted in decreased Wnt signaling and suppressed stem cell activity. The decrease in stem cell expansion was found to be linked to cholinergic signaling. In the mouse stomach, cholinergic nerves regulate gastrointestinal epithelial proliferation [136]. Such denervation has been tested as a therapeutic strategy to treat gastric cancer where it can be used in conjunction with chemotherapy [135]. Nitric oxide (NO) is able to initiate angiogenesis through the generation of eNOS and hydrogen sulfide (H2S). H2S is generated via the action of cystathionine-γ-lyase (CSE) [137].

8. Therapeutic Targeting of the Interplay between Angiogenesis and Neurogenesis

Targeting angiogenesis for therapy in cancer is a well-established strategy that involves targeting the communication between tumor cells and the nearby blood vessels. It is known that interfering with neurogenic signaling can affect cancer development and progression. In mouse and tissue culture models of both prostate and lung cancer, it has been shown that chemical (6-hydroxydopamine, 6-OHDA) and surgical (hypogastric nerve cut) sympathectomy can prevent the development and progression of these cancers [5,73]. Since it appears that angiogenesis is triggered by axonogenesis and neurogenesis through adrenergic signaling, targeting the adrenergic receptor or ligands is a viable treatment option. For instance, perineural invasion (PNI) is a promising therapeutic target. PNI is induced by the activation of the β2-adrenergic receptor, leading to PKA/STAT3 activation, which, in turn, activates NGF, MMP2, and MMP9 expression. Ligands which bind to and activate this receptor in PNI include sympathetic fiber-derived noradrenaline. This can be achieved using drugs, such as propranolol and penbutolol, which are β Adrenergic blockers, or atropine and hyoscine, which are muscarinic antagonists. Studies have shown that these drugs prevent prostate cancer cell migration [138]. Dopamine receptors are also potential drug targets. In mouse models of lung cancer, Dopamine receptor D2 (D2R) agonist inhibits angiogenesis [91]. Dopamine (DA) inhibitors can prevent cancer cell proliferation. These inhibitors lead to the downregulation of ERK1/2 and PI3K/AKT pathways [139]. Both NGF and BDNF rely on Trk receptors to initiate the signaling pathways. The antagonism or inhibition of these Trks could potentially prevent neurotrophin signaling in cancer progression and initiation. This can be used to treat neuropathic pain related to cancer as well as inhibiting neurogenesis, angiogenesis, and their interplay. The Trk receptor inhibitors Larotrectinib and entrectinib have both been approved for use in the treatment of tumors [140]. It has long been known that innervation of gastric tissue promotes the development of gastric cancers. The severing of the vagus nerve at particular branches has been found to inhibit the development of gastric cancers in a mouse model. In addition to this, similar results were achieved when the mice were treated with BOTOX, which acted to block neural signaling [135].

9. Conclusions

The management and treatment of cancer require the advancement of knowledge concerning how tumors interact with their microenvironment. While angiogenesis has been known to contribute to the development and progression of cancer, it is now known that in some solid tumors, infiltrating nerves and catecholaminergic signaling may play an important role in tumor initiation and progression. Angiogenesis and neurogenesis share many of the same signaling pathways and both have a requirement for the rearrangement of tissue as well as the migration of cells. This provides the basis for the interplay between them. The two processes, angiogenesis and neurogenesis, are both activated by cancer cells and share many of the signaling pathways and molecules. This means that both processes are activated in a similar way and can activate each other. This also explains how cancer cells can take advantage of this dual activation to proliferate, migrate, and metastasize. This also makes this interplay an attractive target for the development of new therapies. These therapies can target multiple pathways while only targeting a few components that are shared between these processes (Figure 7). Although not covered in this review in detail, the interplay and signaling pathways involved in these processes also suppress the immune system. This may enable therapies targeting individual molecules to target three hallmarks of cancer. Much of our knowledge on the interplay between these processes comes from animal models of stroke and neurodegenerative disorders. It would be useful if future studies on the role and interplay of these processes in cancer are performed using cancer-specific models. Further knowledge of the interplay between neurogenesis, angiogenesis, nerve–cancer crosstalk, and the neuro–immune axis is required for the implementation of use of anti-neurogenic and anti-angiogenic targets in the treatment of cancer.

Author Contributions

Conceptualization, Z.D. and R.H.; resources, Z.D.; writing—original draft preparation, R.H. and B.P.D.; writing—review and editing, B.P.D., R.K., T.M., Z.D. and D.O.B. All authors have read and agreed to the published version of the manuscript.

Funding

We would like to thank the South African Medical Research Council (SAMRC), Grant Number 23108, and the National Research Foundation (NRF), Grant Number 138139, for funding this work.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Cao, Y.; Arbiser, J.; D’Amato, R.J.; D’Amore, P.A.; Ingber, D.E.; Kerbel, R.; Klagsbrun, M.; Lim, S.; Moses, M.A.; Zetter, B.; et al. Forty-year journey of angiogenesis translational research. Sci. Transl. Med. 2011, 3, 114rv113. [Google Scholar] [CrossRef] [PubMed]
  2. Demir, I.E.; Friess, H.; Ceyhan, G.O. Nerve-cancer interactions in the stromal biology of pancreatic cancer. Front. Physiol. 2012, 3, 97. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Deng, J.; You, Q.; Gao, Y.; Yu, Q.; Zhao, P.; Zheng, Y.; Fang, W.; Xu, N.; Teng, L. Prognostic value of perineural invasion in gastric cancer: A systematic review and meta-analysis. PLoS ONE 2014, 9, e88907. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Schmitd, L.B.; Scanlon, C.S.; D’Silva, N.J. Perineural Invasion in Head and Neck Cancer. J. Dent. Res. 2018, 97, 742–750. [Google Scholar] [CrossRef]
  5. Magnon, C.; Hall, S.J.; Lin, J.; Xue, X.; Gerber, L.; Freedland, S.J.; Frenette, P.S. Autonomic nerve development contributes to prostate cancer progression. Science 2013, 341, 1236361. [Google Scholar] [CrossRef] [Green Version]
  6. Ayala, G.E.; Dai, H.; Powell, M.; Li, R.; Ding, Y.; Wheeler, T.M.; Shine, D.; Kadmon, D.; Thompson, T.; Miles, B.J.; et al. Cancer-related axonogenesis and neurogenesis in prostate cancer. Clin. Cancer Res. 2008, 14, 7593–7603. [Google Scholar] [CrossRef] [Green Version]
  7. Bapat, A.A.; Hostetter, G.; Von Hoff, D.D.; Han, H. Perineural invasion and associated pain in pancreatic cancer. Nat. Rev. Cancer 2011, 11, 695–707. [Google Scholar] [CrossRef]
  8. Carmeliet, P.; Tessier-Lavigne, M. Common mechanisms of nerve and blood vessel wiring. Nature 2005, 436, 193–200. [Google Scholar] [CrossRef]
  9. Arvanitis, C.D.; Ferraro, G.B.; Jain, R.K. The blood-brain barrier and blood-tumour barrier in brain tumours and metastases. Nat. Rev. Cancer 2020, 20, 26–41. [Google Scholar] [CrossRef]
  10. Font, M.A.; Arboix, A.; Krupinski, J. Angiogenesis, neurogenesis and neuroplasticity in ischemic stroke. Curr. Cardiol. Rev. 2010, 6, 238–244. [Google Scholar] [CrossRef] [Green Version]
  11. Dantzer, R. Neuroimmune Interactions: From the Brain to the Immune System and Vice Versa. Physiol. Rev. 2018, 98, 477–504. [Google Scholar] [CrossRef]
  12. Eisenberg, E.; Suzan, E. Drug combinations in the treatment of neuropathic pain. Curr. Pain Headache Rep. 2014, 18, 463. [Google Scholar] [CrossRef]
  13. Slavik, E.; Ivanović, S.; Grujicić, D. Cancer pain (classification and pain syndromes). Acta Chir. Iugosl. 2004, 51, 9–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Schmidt, B.L. The neurobiology of cancer pain. Neuroscientist 2014, 20, 546–562. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Kopp, H.G.; Ramos, C.A.; Rafii, S. Contribution of endothelial progenitors and proangiogenic hematopoietic cells to vascularization of tumor and ischemic tissue. Curr. Opin. Hematol. 2006, 13, 175–181. [Google Scholar] [CrossRef] [PubMed]
  16. Scheau, C.; Badarau, I.A.; Costache, R.; Caruntu, C.; Mihai, G.L.; Didilescu, A.C.; Constantin, C.; Neagu, M. The Role of Matrix Metalloproteinases in the Epithelial-Mesenchymal Transition of Hepatocellular Carcinoma. Anal. Cell. Pathol. 2019, 2019, 9423907. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Jain, R.K. Molecular regulation of vessel maturation. Nat. Med. 2003, 9, 685–693. [Google Scholar] [CrossRef] [PubMed]
  18. Luo, Z.; Shang, X.; Zhang, H.; Wang, G.; Massey, P.A.; Barton, S.R.; Kevil, C.G.; Dong, Y. Notch Signaling in Osteogenesis, Osteoclastogenesis, and Angiogenesis. Am. J. Pathol. 2019, 189, 1495–1500. [Google Scholar] [CrossRef] [Green Version]
  19. Wong, P.P.; Bodrug, N.; Hodivala-Dilke, K.M. Exploring Novel Methods for Modulating Tumor Blood Vessels in Cancer Treatment. Curr. Biol. 2016, 26, R1161–R1166. [Google Scholar] [CrossRef]
  20. Carmeliet, P.; Jain, R.K. Molecular mechanisms and clinical applications of angiogenesis. Nature 2011, 473, 298–307. [Google Scholar] [CrossRef] [Green Version]
  21. Lee, H.W.; Xu, Y.; He, L.; Choi, W.; Gonzalez, D.; Jin, S.W.; Simons, M. Role of Venous Endothelial Cells in Developmental and Pathologic Angiogenesis. Circulation 2021, 144, 1308–1322. [Google Scholar] [CrossRef] [PubMed]
  22. Baeriswyl, V.; Christofori, G. The angiogenic switch in carcinogenesis. Semin. Cancer Biol. 2009, 19, 329–337. [Google Scholar] [CrossRef]
  23. Chen, H.; Liu, D.; Yang, Z.; Sun, L.; Deng, Q.; Yang, S.; Qian, L.; Guo, L.; Yu, M.; Hu, M.; et al. Adrenergic signaling promotes angiogenesis through endothelial cell-tumor cell crosstalk. Endocr. -Relat. Cancer 2014, 21, 783–795. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Huang, T.; Tworoger, S.S.; Hecht, J.L.; Rice, M.S.; Sood, A.K.; Kubzansky, L.D.; Poole, E.M. Association of Ovarian Tumor β2-Adrenergic Receptor Status with Ovarian Cancer Risk Factors and Survival. Cancer Epidemiol. Biomark. Prev. 2016, 25, 1587–1594. [Google Scholar] [CrossRef] [Green Version]
  25. Gong, C.; Hu, B.; Chen, H.; Zhu, J.; Nie, J.; Hua, L.; Chen, L.; Fang, Y.; Hang, C.; Lu, Y. β2-adrenergic receptor drives the metastasis and invasion of pancreatic ductal adenocarcinoma through activating Cdc42 signaling pathway. J. Mol. Histol. 2022, 53, 645–655. [Google Scholar] [CrossRef] [PubMed]
  26. Ray, R.; Al Khashali, H.; Haddad, B.; Wareham, J.; Coleman, K.L.; Alomari, D.; Ranzenberger, R.; Guthrie, J.; Heyl, D.; Evans, H.G. Regulation of Cisplatin Resistance in Lung Cancer Cells by Nicotine, BDNF, and a β-Adrenergic Receptor Blocker. Int. J. Mol. Sci. 2022, 23, 12829. [Google Scholar] [CrossRef]
  27. Encinas, J.M.; Michurina, T.V.; Peunova, N.; Park, J.-H.; Tordo, J.; Peterson, D.A.; Fishell, G.; Koulakov, A.; Enikolopov, G. Division-Coupled Astrocytic Differentiation and Age-Related Depletion of Neural Stem Cells in the Adult Hippocampus. Cell Stem Cell 2011, 8, 566–579. [Google Scholar] [CrossRef] [Green Version]
  28. Katsimpardi, L.; Litterman, N.K.; Schein, P.A.; Miller, C.M.; Loffredo, F.S.; Wojtkiewicz, G.R.; Chen, J.W.; Lee, R.T.; Wagers, A.J.; Rubin, L.L. Vascular and neurogenic rejuvenation of the aging mouse brain by young systemic factors. Science 2014, 344, 630–634. [Google Scholar] [CrossRef] [Green Version]
  29. Ponti, G.; Obernier, K.; Alvarez-Buylla, A. Lineage progression from stem cells to new neurons in the adult brain ventricular-subventricular zone. Cell Cycle 2013, 12, 1649–1650. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Bonaguidi, M.A.; Wheeler, M.A.; Shapiro, J.S.; Stadel, R.P.; Sun, G.J.; Ming, G.-L.; Song, H. In Vivo Clonal Analysis Reveals Self-Renewing and Multipotent Adult Neural Stem Cell Characteristics. Cell 2011, 145, 1142–1155. [Google Scholar] [CrossRef] [Green Version]
  31. Livneh, Y.; Adam, Y.; Mizrahi, A. Odor Processing by Adult-Born Neurons. Neuron 2014, 81, 1097–1110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Fuentealba, L.C.; Obernier, K.; Alvarez-Buylla, A. Adult Neural Stem Cells Bridge Their Niche. Cell Stem Cell 2012, 10, 698–708. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Dranovsky, A.; Picchini, A.M.; Moadel, T.; Sisti, A.C.; Yamada, A.; Kimura, S.; Leonardo, E.D.; Hen, R. Experience Dictates Stem Cell Fate in the Adult Hippocampus. Neuron 2011, 70, 908–923. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Goshen, I.; Kreisel, T.; Ben-Menachem-Zidon, O.; Licht, T.; Weidenfeld, J.; Ben-Hur, T.; Yirmiya, R. Brain interleukin-1 mediates chronic stress-induced depression in mice via adrenocortical activation and hippocampal neurogenesis suppression. Mol. Psychiatry 2008, 13, 717–728. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Thaker, P.H.; Sood, A.K. Neuroendocrine influences on cancer biology. Semin. Cancer Biol. 2008, 18, 164–170. [Google Scholar] [CrossRef] [Green Version]
  36. Armaiz-Pena, G.N.; Lutgendorf, S.K.; Cole, S.W.; Sood, A.K. Neuroendocrine modulation of cancer progression. Brain Behav. Immun. 2009, 23, 10–15. [Google Scholar] [CrossRef] [Green Version]
  37. Marchesi, F.; Piemonti, L.; Mantovani, A.; Allavena, P. Molecular mechanisms of perineural invasion, a forgotten pathway of dissemination and metastasis. Cytokine Growth Factor Rev. 2010, 21, 77–82. [Google Scholar] [CrossRef]
  38. Silverman, D.A.; Martinez, V.K.; Dougherty, P.M.; Myers, J.N.; Calin, G.A.; Amit, M. Cancer-Associated Neurogenesis and Nerve-Cancer Cross-talk. Cancer Res. 2021, 81, 1431–1440. [Google Scholar] [CrossRef]
  39. Ding, L.; Saunders, T.L.; Enikolopov, G.; Morrison, S.J. Endothelial and perivascular cells maintain haematopoietic stem cells. Nature 2012, 481, 457–462. [Google Scholar] [CrossRef] [Green Version]
  40. Mirzadeh, Z.; Merkle, F.T.; Soriano-Navarro, M.; Garcia-Verdugo, J.M.; Alvarez-Buylla, A. Neural Stem Cells Confer Unique Pinwheel Architecture to the Ventricular Surface in Neurogenic Regions of the Adult Brain. Cell Stem Cell 2008, 3, 265–278. [Google Scholar] [CrossRef] [Green Version]
  41. Shen, Q.; Wang, Y.; Kokovay, E.; Lin, G.; Chuang, S.-M.; Goderie, S.K.; Roysam, B.; Temple, S. Adult SVZ Stem Cells Lie in a Vascular Niche: A Quantitative Analysis of Niche Cell-Cell Interactions. Cell Stem Cell 2008, 3, 289–300. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Tavazoie, M.; Van der Veken, L.; Silva-Vargas, V.; Louissaint, M.; Colonna, L.; Zaidi, B.; Garcia-Verdugo, J.M.; Doetsch, F. A Specialized Vascular Niche for Adult Neural Stem Cells. Cell Stem Cell 2008, 3, 279–288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Mauffrey, P.; Tchitchek, N.; Barroca, V.; Bemelmans, A.P.; Firlej, V.; Allory, Y.; Roméo, P.H.; Magnon, C. Progenitors from the central nervous system drive neurogenesis in cancer. Nature 2019, 569, 672–678. [Google Scholar] [CrossRef] [PubMed]
  44. Lu, R.; Fan, C.; Shangguan, W.; Liu, Y.; Li, Y.; Shang, Y.; Yin, D.; Zhang, S.; Huang, Q.; Li, X.; et al. Neurons generated from carcinoma stem cells support cancer progression. Signal Transduct. Target. Ther. 2017, 2, 16036. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Kazanis, I.; Lathia, J.D.; Vadakkan, T.J.; Raborn, E.; Wan, R.; Mughal, M.R.; Eckley, D.M.; Sasaki, T.; Patton, B.; Mattson, M.P.; et al. Quiescence and activation of stem and precursor cell populations in the subependymal zone of the mammalian brain are associated with distinct cellular and extracellular matrix signals. J. Neurosci. 2010, 30, 9771–9781. [Google Scholar] [CrossRef] [Green Version]
  46. Kokovay, E.; Goderie, S.; Wang, Y.; Lotz, S.; Lin, G.; Sun, Y.; Roysam, B.; Shen, Q.; Temple, S. Adult SVZ Lineage Cells Home to and Leave the Vascular Niche via Differential Responses to SDF1/CXCR4 Signaling. Cell Stem Cell 2010, 7, 163–173. [Google Scholar] [CrossRef] [Green Version]
  47. Codega, P.; Silva-Vargas, V.; Paul, A.; Maldonado-Soto, A.R.; DeLeo, A.M.; Pastrana, E.; Doetsch, F. Prospective Identification and Purification of Quiescent Adult Neural Stem Cells from Their In Vivo Niche. Neuron 2014, 82, 545–559. [Google Scholar] [CrossRef] [Green Version]
  48. Delgado, A.C.; Ferrón, S.R.; Vicente, D.; Porlan, E.; Perez-Villalba, A.; Trujillo, C.M.; D′ocón, P.; Fariñas, I. Endothelial NT-3 Delivered by Vasculature and CSF Promotes Quiescence of Subependymal Neural Stem Cells through Nitric Oxide Induction. Neuron 2014, 83, 572–585. [Google Scholar] [CrossRef] [Green Version]
  49. Zhang, R.L.; Chopp, M.; Roberts, C.; Liu, X.; Wei, M.; Nejad-Davarani, S.P.; Wang, X.; Zhang, Z.G. Stroke increases neural stem cells and angiogenesis in the neurogenic niche of the adult mouse. PLoS ONE 2014, 9, e113972. [Google Scholar] [CrossRef] [Green Version]
  50. Teng, H.; Zhang, Z.G.; Wang, L.; Zhang, R.L.; Zhang, L.; Morris, D.; Gregg, S.R.; Wu, Z.; Jiang, A.; Lu, M.; et al. Coupling of angiogenesis and neurogenesis in cultured endothelial cells and neural progenitor cells after stroke. J. Cereb. Blood Flow Metab. 2008, 28, 764–771. [Google Scholar] [CrossRef]
  51. Asgari Taei, A.; Nasoohi, S.; Hassanzadeh, G.; Kadivar, M.; Dargahi, L.; Farahmandfar, M. Enhancement of angiogenesis and neurogenesis by intracerebroventricular injection of secretome from human embryonic stem cell-derived mesenchymal stem cells in ischemic stroke model. Biomed. Pharmacother. Biomed. Pharmacother. 2021, 140, 111709. [Google Scholar] [CrossRef]
  52. Morimoto, T.; Yasuhara, T.; Kameda, M.; Baba, T.; Kuramoto, S.; Kondo, A.; Takahashi, K.; Tajiri, N.; Wang, F.; Meng, J.; et al. Striatal stimulation nurtures endogenous neurogenesis and angiogenesis in chronic-phase ischemic stroke rats. Cell Transplant. 2011, 20, 1049–1064. [Google Scholar] [CrossRef]
  53. Martončíková, M.; Matiašová, A.A.; Ševc, J.; Račeková, E. Relationship between Blood Vessels and Migration of Neuroblasts in the Olfactory Neurogenic Region of the Rodent Brain. Int. J. Mol. Sci. 2021, 22, 11506. [Google Scholar] [CrossRef] [PubMed]
  54. Madelaine, R.; Sloan, S.A.; Huber, N.; Notwell, J.H.; Leung, L.C.; Skariah, G.; Halluin, C.; Paşca, S.P.; Bejerano, G.; Krasnow, M.A.; et al. MicroRNA-9 Couples Brain Neurogenesis and Angiogenesis. Cell Rep. 2017, 20, 1533–1542. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Qu, M.; Pan, J.; Wang, L.; Zhou, P.; Song, Y.; Wang, S.; Jiang, L.; Geng, J.; Zhang, Z.; Wang, Y.; et al. MicroRNA-126 Regulates Angiogenesis and Neurogenesis in a Mouse Model of Focal Cerebral Ischemia. Mol. Therapy. Nucleic Acids 2019, 16, 15–25. [Google Scholar] [CrossRef] [Green Version]
  56. Chen, D.; Wei, L.; Liu, Z.R.; Yang, J.J.; Gu, X.; Wei, Z.Z.; Liu, L.P.; Yu, S.P. Pyruvate Kinase M2 Increases Angiogenesis, Neurogenesis, and Functional Recovery Mediated by Upregulation of STAT3 and Focal Adhesion Kinase Activities After Ischemic Stroke in Adult Mice. Neurotherapeutics 2018, 15, 770–784. [Google Scholar] [CrossRef] [Green Version]
  57. Jin, Y.; Barnett, A.; Zhang, Y.; Yu, X.; Luo, Y. Poststroke Sonic Hedgehog Agonist Treatment Improves Functional Recovery by Enhancing Neurogenesis and Angiogenesis. Stroke 2017, 48, 1636–1645. [Google Scholar] [CrossRef] [PubMed]
  58. Jing, X.; Miwa, H.; Sawada, T.; Nakanishi, I.; Kondo, T.; Miyajima, M.; Sakaguchi, K. Ephrin-A1-mediated dopaminergic neurogenesis and angiogenesis in a rat model of Parkinson’s disease. PLoS ONE 2012, 7, e32019. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Ziani, L.; Chouaib, S.; Thiery, J. Alteration of the Antitumor Immune Response by Cancer-Associated Fibroblasts. Front. Immunol. 2018, 9, 414. [Google Scholar] [CrossRef] [PubMed]
  60. Vázquez-Prado, J.; Bracho-Valdés, I.; Cervantes-Villagrana, R.D.; Reyes-Cruz, G. Gβγ Pathways in Cell Polarity and Migration Linked to Oncogenic GPCR Signaling: Potential Relevance in Tumor Microenvironment. Mol. Pharmacol. 2016, 90, 573–586. [Google Scholar] [CrossRef] [Green Version]
  61. Venneri, M.A.; De Palma, M.; Ponzoni, M.; Pucci, F.; Scielzo, C.; Zonari, E.; Mazzieri, R.; Doglioni, C.; Naldini, L. Identification of proangiogenic TIE2-expressing monocytes (TEMs) in human peripheral blood and cancer. Blood 2007, 109, 5276–5285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Li, F.; Zhao, Y.; Wei, L.; Li, S.; Liu, J. Tumor-infiltrating Treg, MDSC, and IDO expression associated with outcomes of neoadjuvant chemotherapy of breast cancer. Cancer Biol. Ther. 2018, 19, 695–705. [Google Scholar] [CrossRef] [Green Version]
  63. Saloman, J.L.; Albers, K.M.; Rhim, A.D.; Davis, B.M. Can Stopping Nerves, Stop Cancer? Trends Neurosci. 2016, 39, 880–889. [Google Scholar] [CrossRef] [Green Version]
  64. Talmadge, J.E.; Fidler, I.J. AACR centennial series: The biology of cancer metastasis: Historical perspective. Cancer Res. 2010, 70, 5649–5669. [Google Scholar] [CrossRef] [Green Version]
  65. Amit, M.; Na’ara, S.; Gil, Z. Mechanisms of cancer dissemination along nerves. Nat. Rev. Cancer 2016, 16, 399–408. [Google Scholar] [CrossRef]
  66. Da Cunha, B.R.; Domingos, C.; Stefanini, A.C.B.; Henrique, T.; Polachini, G.M.; Castelo-Branco, P.; Tajara, E.H. Cellular Interactions in the Tumor Microenvironment: The Role of Secretome. J. Cancer 2019, 10, 4574–4587. [Google Scholar] [CrossRef] [Green Version]
  67. Mantyh, P.W.; Koltzenburg, M.; Mendell, L.M.; Tive, L.; Shelton, D.L. Antagonism of nerve growth factor-TrkA signaling and the relief of pain. Anesthesiology 2011, 115, 189–204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Kusakabe, T.; Sawaji, Y.; Endo, K.; Suzuki, H.; Konishi, T.; Maekawa, A.; Murata, K.; Yamamoto, K. DUSP-1 Induced by PGE(2) and PGE(1) Attenuates IL-1β-Activated MAPK Signaling, Leading to Suppression of NGF Expression in Human Intervertebral Disc Cells. Int. J. Mol. Sci. 2021, 23, 371. [Google Scholar] [CrossRef]
  69. Jimenez-Andrade, J.M.; Mantyh, P.W. Sensory and sympathetic nerve fibers undergo sprouting and neuroma formation in the painful arthritic joint of geriatric mice. Arthritis Res. Ther. 2012, 14, R101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Zhang, R.L.; Zhang, Z.G.; Lu, M.; Wang, Y.; Yang, J.J.; Chopp, M. Reduction of the cell cycle length by decreasing G1 phase and cell cycle reentry expand neuronal progenitor cells in the subventricular zone of adult rat after stroke. J. Cereb. Blood Flow Metab. 2006, 26, 857–863. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Götz, M.; Huttner, W.B. The cell biology of neurogenesis. Nat. Rev. Mol. Cell Biol. 2005, 6, 777–788. [Google Scholar] [CrossRef]
  72. Zahalka, A.H.; Arnal-Estapé, A.; Maryanovich, M.; Nakahara, F.; Cruz, C.D.; Finley, L.W.S.; Frenette, P.S. Adrenergic nerves activate an angio-metabolic switch in prostate cancer. Science 2017, 358, 321–326. [Google Scholar] [CrossRef] [Green Version]
  73. Xia, Y.; Wei, Y.; Li, Z.Y.; Cai, X.Y.; Zhang, L.L.; Dong, X.R.; Zhang, S.; Zhang, R.G.; Meng, R.; Zhu, F.; et al. Catecholamines contribute to the neovascularization of lung cancer via tumor-associated macrophages. Brain Behav. Immun. 2019, 81, 111–121. [Google Scholar] [CrossRef] [PubMed]
  74. Yamashita, T.; Ninomiya, M.; Acosta, P.H.; García-Verdugo, J.M.; Sunabori, T.; Sakaguchi, M.; Adachi, K.; Kojima, T.; Hirota, Y.; Kawase, T.; et al. Subventricular zone-derived neuroblasts migrate and differentiate into mature neurons in the post-stroke adult striatum. J. Neurosci. 2006, 26, 6627–6636. [Google Scholar] [CrossRef] [Green Version]
  75. Sawada, A.; Niiyama, Y.; Ataka, K.; Nagaishi, K.; Yamakage, M.; Fujimiya, M. Suppression of bone marrow-derived microglia in the amygdala improves anxiety-like behavior induced by chronic partial sciatic nerve ligation in mice. Pain 2014, 155, 1762–1772. [Google Scholar] [CrossRef] [PubMed]
  76. Hattori, F.; Murayama, N.; Noshita, T.; Oikawa, S. Mitochondrial peroxiredoxin-3 protects hippocampal neurons from excitotoxic injury in vivo. J. Neurochem. 2003, 86, 860–868. [Google Scholar] [CrossRef]
  77. Robin, M.; Andreu-Gallien, J.; Schlageter, M.H.; Bengoufa, D.; Guillemot, I.; Pokorna, K.; Robert, C.; Larghero, J.; Rousselot, P.; Raffoux, E.; et al. Frequent antibody production against RARalpha in both APL mice and patients. Blood 2006, 108, 1972–1974. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Wang, T.; Choi, E.; Monaco, M.C.; Major, E.O.; Medynets, M.; Nath, A. Direct induction of human neural stem cells from peripheral blood hematopoietic progenitor cells. J. Vis. Exp. 2015, 95, e52298. [Google Scholar] [CrossRef] [Green Version]
  79. Katakowski, M.; Zhang, Z.G.; Chen, J.; Zhang, R.; Wang, Y.; Jiang, H.; Zhang, L.; Robin, A.; Li, Y.; Chopp, M. Phosphoinositide 3-kinase promotes adult subventricular neuroblast migration after stroke. J. Neurosci. Res. 2003, 74, 494–501. [Google Scholar] [CrossRef]
  80. Barkho, B.Z.; Munoz, A.E.; Li, X.; Li, L.; Cunningham, L.A.; Zhao, X. Endogenous matrix metalloproteinase (MMP)-3 and MMP-9 promote the differentiation and migration of adult neural progenitor cells in response to chemokines. Stem Cells 2008, 26, 3139–3149. [Google Scholar] [CrossRef] [Green Version]
  81. Douet, V.; Kerever, A.; Arikawa-Hirasawa, E.; Mercier, F. Fractone-heparan sulphates mediate FGF-2 stimulation of cell proliferation in the adult subventricular zone. Cell Prolif. 2013, 46, 137–145. [Google Scholar] [CrossRef] [PubMed]
  82. Mercier, F.; Douet, V. Bone morphogenetic protein-4 inhibits adult neurogenesis and is regulated by fractone-associated heparan sulfates in the subventricular zone. J. Chem. Neuroanat. 2014, 57–58, 54–61. [Google Scholar] [CrossRef] [PubMed]
  83. Shingo, T.; Gregg, C.; Enwere, E.; Fujikawa, H.; Hassam, R.; Geary, C.; Cross, J.C.; Weiss, S. Pregnancy-stimulated neurogenesis in the adult female forebrain mediated by prolactin. Science 2003, 299, 117–120. [Google Scholar] [CrossRef] [PubMed]
  84. Andreu-Agulló, C.; Morante-Redolat, J.M.; Delgado, A.C.; Fariñas, I. Vascular niche factor PEDF modulates Notch-dependent stemness in the adult subependymal zone. Nat. Neurosci. 2009, 12, 1514–1523. [Google Scholar] [CrossRef] [PubMed]
  85. Mehlen, P.; Delloye-Bourgeois, C.; Chédotal, A. Novel roles for Slits and netrins: Axon guidance cues as anticancer targets? Nat. Rev. Cancer 2011, 11, 188–197. [Google Scholar] [CrossRef]
  86. Sasahira, T.; Ueda, N.; Yamamoto, K.; Bhawal, U.K.; Kurihara, M.; Kirita, T.; Kuniyasu, H. Trks are novel oncogenes involved in the induction of neovascularization, tumor progression, and nodal metastasis in oral squamous cell carcinoma. Clin. Exp. Metastasis 2013, 30, 165–176. [Google Scholar] [CrossRef]
  87. Au, C.W.; Siu, M.K.; Liao, X.; Wong, E.S.; Ngan, H.Y.; Tam, K.F.; Chan, D.C.; Chan, Q.K.; Cheung, A.N. Tyrosine kinase B receptor and BDNF expression in ovarian cancers—Effect on cell migration, angiogenesis and clinical outcome. Cancer Lett. 2009, 281, 151–161. [Google Scholar] [CrossRef]
  88. Lam, C.T.; Yang, Z.F.; Lau, C.K.; Tam, K.H.; Fan, S.T.; Poon, R.T. Brain-derived neurotrophic factor promotes tumorigenesis via induction of neovascularization: Implication in hepatocellular carcinoma. Clin. Cancer Res. 2011, 17, 3123–3133. [Google Scholar] [CrossRef] [Green Version]
  89. Mu, J.; Huang, W.; Tan, Z.; Li, M.; Zhang, L.; Ding, Q.; Wu, X.; Lu, J.; Liu, Y.; Dong, Q.; et al. Dopamine receptor D2 is correlated with gastric cancer prognosis. Oncol. Lett. 2017, 13, 1223–1227. [Google Scholar] [CrossRef] [Green Version]
  90. Mao, J.J.; Saper, R.B.; Chesney, M.A. The role of academic health centres to inform evidence-based integrative oncology practice. Nat. Rev. Cancer 2015, 15, 247. [Google Scholar] [CrossRef] [Green Version]
  91. Hoeppner, L.H.; Sinha, S.; Wang, Y.; Bhattacharya, R.; Dutta, S.; Gong, X.; Bedell, V.M.; Suresh, S.; Chun, C.; Ramchandran, R.; et al. RhoC maintains vascular homeostasis by regulating VEGF-induced signaling in endothelial cells. J. Cell Sci. 2015, 128, 3556–3568. [Google Scholar] [CrossRef] [Green Version]
  92. Akl, M.R.; Nagpal, P.; Ayoub, N.M.; Tai, B.; Prabhu, S.A.; Capac, C.M.; Gliksman, M.; Goy, A.; Suh, K.S. Molecular and clinical significance of fibroblast growth factor 2 (FGF2 /bFGF) in malignancies of solid and hematological cancers for personalized therapies. Oncotarget 2016, 7, 44735–44762. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Yoshimura, S.; Takagi, Y.; Harada, J.; Teramoto, T.; Thomas, S.S.; Waeber, C.; Bakowska, J.C.; Breakefield, X.O.; Moskowitz, M.A. FGF-2 regulation of neurogenesis in adult hippocampus after brain injury. Proc. Natl. Acad. Sci. USA 2001, 98, 5874–5879. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Ikeda, N.; Nonoguchi, N.; Zhao, M.Z.; Watanabe, T.; Kajimoto, Y.; Furutama, D.; Kimura, F.; Dezawa, M.; Coffin, R.S.; Otsuki, Y.; et al. Bone marrow stromal cells that enhanced fibroblast growth factor-2 secretion by herpes simplex virus vector improve neurological outcome after transient focal cerebral ischemia in rats. Stroke 2005, 36, 2725–2730. [Google Scholar] [CrossRef] [Green Version]
  95. Billottet, C.; Janji, B.; Thiery, J.P.; Jouanneau, J. Rapid tumor development and potent vascularization are independent events in carcinoma producing FGF-1 or FGF-2. Oncogene 2002, 21, 8128–8139. [Google Scholar] [CrossRef] [Green Version]
  96. Taraboletti, G.; D’Ascenzo, S.; Borsotti, P.; Giavazzi, R.; Pavan, A.; Dolo, V. Shedding of the matrix metalloproteinases MMP-2, MMP-9, and MT1-MMP as membrane vesicle-associated components by endothelial cells. Am. J. Pathol. 2002, 160, 673–680. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Kalluri, H.S.; Vemuganti, R.; Dempsey, R.J. Mechanism of insulin-like growth factor I-mediated proliferation of adult neural progenitor cells: Role of Akt. Eur. J. Neurosci. 2007, 25, 1041–1048. [Google Scholar] [CrossRef]
  98. Zhang, Z.G.; Chopp, M. Neurorestorative therapies for stroke: Underlying mechanisms and translation to the clinic. Lancet. Neurol. 2009, 8, 491–500. [Google Scholar] [CrossRef] [Green Version]
  99. Jacobo, S.M.; Kazlauskas, A. Insulin-like growth factor 1 (IGF-1) stabilizes nascent blood vessels. J. Biol. Chem. 2015, 290, 6349–6360. [Google Scholar] [CrossRef] [Green Version]
  100. Carmeliet, P.; Storkebaum, E. Vascular and neuronal effects of VEGF in the nervous system: Implications for neurological disorders. Semin. Cell Dev. Biol. 2002, 13, 39–53. [Google Scholar] [CrossRef]
  101. Maurer, M.H.; Tripps, W.K.; Feldmann, R.E., Jr.; Kuschinsky, W. Expression of vascular endothelial growth factor and its receptors in rat neural stem cells. Neurosci. Lett. 2003, 344, 165–168. [Google Scholar] [CrossRef]
  102. Scharfman, H.; Goodman, J.; Macleod, A.; Phani, S.; Antonelli, C.; Croll, S. Increased neurogenesis and the ectopic granule cells after intrahippocampal BDNF infusion in adult rats. Exp. Neurol. 2005, 192, 348–356. [Google Scholar] [CrossRef]
  103. Andsberg, G.; Kokaia, Z.; Klein, R.L.; Muzyczka, N.; Lindvall, O.; Mandel, R.J. Neuropathological and behavioral consequences of adeno-associated viral vector-mediated continuous intrastriatal neurotrophin delivery in a focal ischemia model in rats. Neurobiol. Dis. 2002, 9, 187–204. [Google Scholar] [CrossRef] [Green Version]
  104. Halade, G.V.; Ma, Y.; Ramirez, T.A.; Zhang, J.; Dai, Q.; Hensler, J.G.; Lopez, E.F.; Ghasemi, O.; Jin, Y.F.; Lindsey, M.L. Reduced BDNF attenuates inflammation and angiogenesis to improve survival and cardiac function following myocardial infarction in mice. Am. J. Physiol. Heart Circ. Physiol. 2013, 305, H1830–H1842. [Google Scholar] [CrossRef] [Green Version]
  105. Descamps, B.; Saif, J.; Benest, A.V.; Biglino, G.; Bates, D.O.; Chamorro-Jorganes, A.; Emanueli, C. BDNF (Brain-Derived Neurotrophic Factor) Promotes Embryonic Stem Cells Differentiation to Endothelial Cells Via a Molecular Pathway, Including MicroRNA-214, EZH2 (Enhancer of Zeste Homolog 2), and eNOS (Endothelial Nitric Oxide Synthase). Arterioscler. Thromb. Vasc. Biol. 2018, 38, 2117–2125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. 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] [PubMed]
  107. Dollé, J.P.; Rezvan, A.; Allen, F.D.; Lazarovici, P.; Lelkes, P.I. Nerve growth factor-induced migration of endothelial cells. J. Pharmacol. Exp. Ther. 2005, 315, 1220–1227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Lazarovici, P.; Gazit, A.; Staniszewska, I.; Marcinkiewicz, C.; Lelkes, P.I. Nerve growth factor (NGF) promotes angiogenesis in the quail chorioallantoic membrane. Endothelium 2006, 13, 51–59. [Google Scholar] [CrossRef]
  109. Julio-Pieper, M.; Lozada, P.; Tapia, V.; Vega, M.; Miranda, C.; Vantman, D.; Ojeda, S.R.; Romero, C. Nerve growth factor induces vascular endothelial growth factor expression in granulosa cells via a trkA receptor/mitogen-activated protein kinase-extracellularly regulated kinase 2-dependent pathway. J. Clin. Endocrinol. Metab. 2009, 94, 3065–3071. [Google Scholar] [CrossRef] [Green Version]
  110. Krupkova, O., Jr.; Loja, T.; Zambo, I.; Veselska, R. Nestin expression in human tumors and tumor cell lines. Neoplasma 2010, 57, 291–298. [Google Scholar] [CrossRef]
  111. Bagri, A.; Tessier-Lavigne, M.; Watts, R.J. Neuropilins in tumor biology. Clin. Cancer 2009, 15, 1860–1864. [Google Scholar] [CrossRef] [Green Version]
  112. Sarabipour, S.; Mac Gabhann, F. VEGF-A121a binding to Neuropilins—A concept revisited. Cell Adhes. Migr. 2018, 12, 204–214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Tata, M.; Ruhrberg, C.; Fantin, A. Vascularisation of the central nervous system. Mech. Dev. 2015, 138 Pt 1, 26–36. [Google Scholar] [CrossRef] [PubMed]
  114. Roy, S.; Bag, A.K.; Singh, R.K.; Talmadge, J.E.; Batra, S.K.; Datta, K. Multifaceted Role of Neuropilins in the Immune System: Potential Targets for Immunotherapy. Front. Immunol. 2017, 8, 1228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Mecollari, V.; Nieuwenhuis, B.; Verhaagen, J. A perspective on the role of class III semaphorin signaling in central nervous system trauma. Front. Cell. Neurosci. 2014, 8, 328. [Google Scholar] [CrossRef] [Green Version]
  116. Hamerlik, P.; Lathia, J.D.; Rasmussen, R.; Wu, Q.; Bartkova, J.; Lee, M.; Moudry, P.; Bartek, J., Jr.; Fischer, W.; Lukas, J.; et al. Autocrine VEGF-VEGFR2-Neuropilin-1 signaling promotes glioma stem-like cell viability and tumor growth. J. Exp. Med. 2012, 209, 507–520. [Google Scholar] [CrossRef] [Green Version]
  117. Bagci, T.; Wu, J.K.; Pfannl, R.; Ilag, L.L.; Jay, D.G. Autocrine semaphorin 3A signaling promotes glioblastoma dispersal. Oncogene 2009, 28, 3537–3550. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Rizzolio, S.; Tamagnone, L. Multifaceted role of neuropilins in cancer. Curr. Med. Chem. 2011, 18, 3563–3575. [Google Scholar] [CrossRef]
  119. Rizzolio, S.; Rabinowicz, N.; Rainero, E.; Lanzetti, L.; Serini, G.; Norman, J.; Neufeld, G.; Tamagnone, L. Neuropilin-1-dependent regulation of EGF-receptor signaling. Cancer Res. 2012, 72, 5801–5811. [Google Scholar] [CrossRef] [Green Version]
  120. Wild, J.R.; Staton, C.A.; Chapple, K.; Corfe, B.M. Neuropilins: Expression and roles in the epithelium. Int. J. Exp. Pathol. 2012, 93, 81–103. [Google Scholar] [CrossRef]
  121. Niland, S.; Eble, J.A. Neuropilins in the Context of Tumor Vasculature. Int. J. Mol. Sci. 2019, 20, 639. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Vogelstein, B.; Papadopoulos, N.; Velculescu, V.E.; Zhou, S.; Diaz, L.A., Jr.; Kinzler, K.W. Cancer genome landscapes. Science 2013, 339, 1546–1558. [Google Scholar] [CrossRef]
  123. Gerhardt, H.; Ruhrberg, C.; Abramsson, A.; Fujisawa, H.; Shima, D.; Betsholtz, C. Neuropilin-1 is required for endothelial tip cell guidance in the developing central nervous system. Dev. Dyn. 2004, 231, 503–509. [Google Scholar] [CrossRef] [PubMed]
  124. Fantin, A.; Herzog, B.; Mahmoud, M.; Yamaji, M.; Plein, A.; Denti, L.; Ruhrberg, C.; Zachary, I. Neuropilin 1 (NRP1) hypomorphism combined with defective VEGF-A binding reveals novel roles for NRP1 in developmental and pathological angiogenesis. Development 2014, 141, 556–562. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Schwarz, Q.; Ruhrberg, C. Neuropilin, you gotta let me know: Should I stay or should I go? Cell Adhes. Migr. 2010, 4, 61–66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Cackowski, F.C.; Xu, L.; Hu, B.; Cheng, S.Y. Identification of two novel alternatively spliced Neuropilin-1 isoforms. Genomics 2004, 84, 82–94. [Google Scholar] [CrossRef] [Green Version]
  127. Chu, P.Y.; Koh, A.P.; Antony, J.; Huang, R.Y. Applications of the Chick Chorioallantoic Membrane as an Alternative Model for Cancer Studies. Cells Tissues Organs 2022, 211, 222–237. [Google Scholar] [CrossRef]
  128. De Palma, M.; Venneri, M.A.; Roca, C.; Naldini, L. Targeting exogenous genes to tumor angiogenesis by transplantation of genetically modified hematopoietic stem cells. Nat. Med. 2003, 9, 789–795. [Google Scholar] [CrossRef]
  129. Evangelista, A.M.; Thompson, M.D.; Bolotina, V.M.; Tong, X.; Cohen, R.A. Nox4- and Nox2-dependent oxidant production is required for VEGF-induced SERCA cysteine-674 S-glutathiolation and endothelial cell migration. Free. Radic. Biol. Med. 2012, 53, 2327–2334. [Google Scholar] [CrossRef] [Green Version]
  130. Harrison, I.P.; Vinh, A.; Johnson, I.R.; Luong, R.; Drummond, G.R.; Sobey, C.G.; Tiganis, T.; Williams, E.D.; O’Leary, J.J.; Brooks, D.A.; et al. NOX2 oxidase expressed in endosomes promotes cell proliferation and prostate tumour development. Oncotarget 2018, 9, 35378–35393. [Google Scholar] [CrossRef] [Green Version]
  131. Serrano, F.; Kolluri, N.S.; Wientjes, F.B.; Card, J.P.; Klann, E. NADPH oxidase immunoreactivity in the mouse brain. Brain Res. 2003, 988, 193–198. [Google Scholar] [CrossRef]
  132. Vallet, P.; Charnay, Y.; Steger, K.; Ogier-Denis, E.; Kovari, E.; Herrmann, F.; Michel, J.P.; Szanto, I. Neuronal expression of the NADPH oxidase NOX4, and its regulation in mouse experimental brain ischemia. Neuroscience 2005, 132, 233–238. [Google Scholar] [CrossRef] [PubMed]
  133. Ibi, M.; Katsuyama, M.; Fan, C.; Iwata, K.; Nishinaka, T.; Yokoyama, T.; Yabe-Nishimura, C. NOX1/NADPH oxidase negatively regulates nerve growth factor-induced neurite outgrowth. Free. Radic. Biol. Med. 2006, 40, 1785–1795. [Google Scholar] [CrossRef]
  134. Kim, S.H.; Won, S.J.; Sohn, S.; Kwon, H.J.; Lee, J.Y.; Park, J.H.; Gwag, B.J. Brain-derived neurotrophic factor can act as a pronecrotic factor through transcriptional and translational activation of NADPH oxidase. J. Cell Biol. 2002, 159, 821–831. [Google Scholar] [CrossRef] [PubMed]
  135. Zhao, C.M.; Hayakawa, Y.; Kodama, Y.; Muthupalani, S.; Westphalen, C.B.; Andersen, G.T.; Flatberg, A.; Johannessen, H.; Friedman, R.A.; Renz, B.W.; et al. Denervation suppresses gastric tumorigenesis. Sci. Transl. Med. 2014, 6, 250ra115. [Google Scholar] [CrossRef] [Green Version]
  136. Ferrarelli, L.K. Of nerves and stomach cancer. Sci. Signal. 2017, 10, eaan0914. [Google Scholar] [CrossRef]
  137. Coletta, C.; Papapetropoulos, A.; Erdelyi, K.; Olah, G.; Módis, K.; Panopoulos, P.; Asimakopoulou, A.; Gerö, D.; Sharina, I.; Martin, E.; et al. Hydrogen sulfide and nitric oxide are mutually dependent in the regulation of angiogenesis and endothelium-dependent vasorelaxation. Proc. Natl. Acad. Sci. USA 2012, 109, 9161–9166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Guo, Z.; Liu, D.; Su, Z. CIP2A mediates prostate cancer progression via the c-Myc signaling pathway. Tumour Biol. 2015, 36, 4777–4783. [Google Scholar] [CrossRef]
  139. Gao, J.; Zhang, C.; Gao, F.; Li, H. The effect and mechanism of dopamine D1 receptors on the proliferation of osteosarcoma cells. Mol. Cell. Biochem. 2017, 430, 31–36. [Google Scholar] [CrossRef]
  140. Al-Salama, Z.T.; Syed, Y.Y.; Scott, L.J. Lenvatinib: A Review in Hepatocellular Carcinoma. Drugs 2019, 79, 665–674. [Google Scholar] [CrossRef]
Figure 1. The physiological relationship between the vascular and nervous systems. (A) The vascular and neural systems mirror each other in the body as they both are required to service the entire body (B). The structure of the neurovascular unit in the brain and the blood brain barrier.
Figure 1. The physiological relationship between the vascular and nervous systems. (A) The vascular and neural systems mirror each other in the body as they both are required to service the entire body (B). The structure of the neurovascular unit in the brain and the blood brain barrier.
Cancers 15 01805 g001aCancers 15 01805 g001b
Figure 2. FGF-2 signaling pathways. The binding of FGF-2 to the FGF receptor FGF-R. The FGF-2 signaling pathway activates the Ras/ERK pathway, which leads to the activation of pro-angiogenic and pro-neurogenic genes. ERK—extracellular signal-regulated kinase, FRS2β—fibroblast growth factor (FGF) receptor substrate 2, GRB1/2—Growth factor receptor-bound protein 1/2, MEK—Mitogen-activated protein kinase, PI3K—Phosphoinositide 3-kinase, Rnd1—Rho-related GTP-binding protein, Shp2—Src homology region 2, SOS—Son of Sevenless.
Figure 2. FGF-2 signaling pathways. The binding of FGF-2 to the FGF receptor FGF-R. The FGF-2 signaling pathway activates the Ras/ERK pathway, which leads to the activation of pro-angiogenic and pro-neurogenic genes. ERK—extracellular signal-regulated kinase, FRS2β—fibroblast growth factor (FGF) receptor substrate 2, GRB1/2—Growth factor receptor-bound protein 1/2, MEK—Mitogen-activated protein kinase, PI3K—Phosphoinositide 3-kinase, Rnd1—Rho-related GTP-binding protein, Shp2—Src homology region 2, SOS—Son of Sevenless.
Cancers 15 01805 g002
Figure 3. IGF-1 signaling pathways: The binding of IGF-1 to the IGF receptor FGF-1R. The IGF-1 signaling pathway activates the Ras/ERK pathway, which leads to the activation of pro-angiogenic and pro-neurogenic genes. In addition to this, the pathway activates the Brn-4 transcription factor, which stimulates neuronal growth and differentiation. BRN4—POU domain, class 3, transcription factor 4, eNOS—Endothelial NOS, ERK—extracellular signal-regulated kinase, FRS2β—fibroblast growth factor (FGF) receptor substrate 2, FOXO 1/3a—Forkhead family of transcription factors, GRB1/2—Growth factor receptor-bound protein 1/2, IRS1/2—Insulin receptor substrate 1/2, MEK—Mitogen activated kinase, mTORC—mammalian target of rapamycin complex 1, PDK1—Pyruvate dehydrogenase kinase isozyme 1, PIP-2/3—phospholipase, PI3K—Phosphoinositide 3-kinase, Shp2—Src homology region 2.
Figure 3. IGF-1 signaling pathways: The binding of IGF-1 to the IGF receptor FGF-1R. The IGF-1 signaling pathway activates the Ras/ERK pathway, which leads to the activation of pro-angiogenic and pro-neurogenic genes. In addition to this, the pathway activates the Brn-4 transcription factor, which stimulates neuronal growth and differentiation. BRN4—POU domain, class 3, transcription factor 4, eNOS—Endothelial NOS, ERK—extracellular signal-regulated kinase, FRS2β—fibroblast growth factor (FGF) receptor substrate 2, FOXO 1/3a—Forkhead family of transcription factors, GRB1/2—Growth factor receptor-bound protein 1/2, IRS1/2—Insulin receptor substrate 1/2, MEK—Mitogen activated kinase, mTORC—mammalian target of rapamycin complex 1, PDK1—Pyruvate dehydrogenase kinase isozyme 1, PIP-2/3—phospholipase, PI3K—Phosphoinositide 3-kinase, Shp2—Src homology region 2.
Cancers 15 01805 g003
Figure 4. VEGF signaling: VEGFR-2 activation stimulates migration and extracellular matrix (ECM) invasion. The BEGF pathway results in neurogenesis, which leads, in combination with increased vascular permeability, to increased angiogenesis. CASP9—Caspase9, eNOS—Endothelial NOS, ERK- extracellular signal-regulated kinase, FAK—Protein-tyrosine kinase, FRS2β—fibroblast growth factor (FGF) receptor substrate 2, FOXO 1/3a—Forkhead family of transcription factors, FSP-27—Fat specific protein 27, IRS1/2—Insulin receptor substrate 1/2, MEK-Mitogen activated kinase, NFAT—Nuclear factor of activated T-cells, PIP-2/3-phospholipase, PI3K—Phosphoinositide 3-kinase, PKC—Protein kinase C, PLCγ—Phosphoinositide phospholipase C, Shp2—Src homology region 2, SPK-SR protein kinase.
Figure 4. VEGF signaling: VEGFR-2 activation stimulates migration and extracellular matrix (ECM) invasion. The BEGF pathway results in neurogenesis, which leads, in combination with increased vascular permeability, to increased angiogenesis. CASP9—Caspase9, eNOS—Endothelial NOS, ERK- extracellular signal-regulated kinase, FAK—Protein-tyrosine kinase, FRS2β—fibroblast growth factor (FGF) receptor substrate 2, FOXO 1/3a—Forkhead family of transcription factors, FSP-27—Fat specific protein 27, IRS1/2—Insulin receptor substrate 1/2, MEK-Mitogen activated kinase, NFAT—Nuclear factor of activated T-cells, PIP-2/3-phospholipase, PI3K—Phosphoinositide 3-kinase, PKC—Protein kinase C, PLCγ—Phosphoinositide phospholipase C, Shp2—Src homology region 2, SPK-SR protein kinase.
Cancers 15 01805 g004
Figure 5. BDNF signaling pathways. Binding of BDNF to TrkB leads to the activation of MAPK, PI3K, and PLCγ pathways. The Shc adaptor protein recruits the growth-factor-receptor-bound protein 2 (grb2) to form a complex with Ras. This initiates ERK, which then activates the CREB transcription factor. Binding of TrkB also activates the PI3K and MF-κβ signaling pathways. These pathways play a role in neurotrophic functions (survival, growth, and differentiation). The PLCγ pathway generates inositol-1, 4, 5-triphosphate (IP3) and diacylglycerol (DAG), resulting in Ca2+/CaMKI activation and the initiation of the transcription of genes that are pro-angiogenic and pro-neurogenic. CREB—Cyclic AMP-responsive element-binding protein, ERK—extracellular signal-regulated kinase, GRB1/2—Growth factor receptor-bound protein 1/2, MEK—Mitogen activated kinase, PIP-2/3—phospholipase, PI3K—Phosphoinositide 3-kinase, PKC—Protein kinase C, PLCγ—Phosphoinositide phospholipase C, Shp2—Src homology region 2.
Figure 5. BDNF signaling pathways. Binding of BDNF to TrkB leads to the activation of MAPK, PI3K, and PLCγ pathways. The Shc adaptor protein recruits the growth-factor-receptor-bound protein 2 (grb2) to form a complex with Ras. This initiates ERK, which then activates the CREB transcription factor. Binding of TrkB also activates the PI3K and MF-κβ signaling pathways. These pathways play a role in neurotrophic functions (survival, growth, and differentiation). The PLCγ pathway generates inositol-1, 4, 5-triphosphate (IP3) and diacylglycerol (DAG), resulting in Ca2+/CaMKI activation and the initiation of the transcription of genes that are pro-angiogenic and pro-neurogenic. CREB—Cyclic AMP-responsive element-binding protein, ERK—extracellular signal-regulated kinase, GRB1/2—Growth factor receptor-bound protein 1/2, MEK—Mitogen activated kinase, PIP-2/3—phospholipase, PI3K—Phosphoinositide 3-kinase, PKC—Protein kinase C, PLCγ—Phosphoinositide phospholipase C, Shp2—Src homology region 2.
Cancers 15 01805 g005
Figure 6. NGF signaling pathways. NGF binding to TrkA and p75NTR receptors promotes neuronal growth. The activation of the ERK signaling pathway promotes the transcription of pro-angiogenic and pro-neurogenic genes as in other neurotrophic and growth factor pathways. CREB—Cyclic AMP-responsive element-binding protein, ELK—ETS domain-containing protein, eNOS—Endothelial NOS, ERK—extracellular signal-regulated kinase, GRB1/2—Growth factor receptor-bound protein 1/2, MEK—Mitogen activated kinase, PDK—Pyruvate dehydrogenase kinase 1, PIP-2/3—phospholipase, PI3K—Phosphoinositide 3-kinase, PKC—Protein kinase C, PLCγ—Phosphoinositide phospholipase C, Shp2—Src homology region 2, SOS—Son of Sevenless.
Figure 6. NGF signaling pathways. NGF binding to TrkA and p75NTR receptors promotes neuronal growth. The activation of the ERK signaling pathway promotes the transcription of pro-angiogenic and pro-neurogenic genes as in other neurotrophic and growth factor pathways. CREB—Cyclic AMP-responsive element-binding protein, ELK—ETS domain-containing protein, eNOS—Endothelial NOS, ERK—extracellular signal-regulated kinase, GRB1/2—Growth factor receptor-bound protein 1/2, MEK—Mitogen activated kinase, PDK—Pyruvate dehydrogenase kinase 1, PIP-2/3—phospholipase, PI3K—Phosphoinositide 3-kinase, PKC—Protein kinase C, PLCγ—Phosphoinositide phospholipase C, Shp2—Src homology region 2, SOS—Son of Sevenless.
Cancers 15 01805 g006
Figure 7. Relationship between neurogenesis and angiogenesis in cancer. The interplay between angiogenesis and neurogenesis is due to signaling molecules, such as neurotrophins, neurotransmitters, and growth factors, which all act to stimulate both processes. These processes can also be stimulated by reactive oxygen species. Both the angiogenic and neurogenic signaling pathways also act to suppress the immune system. Apart from cancer, the interplay between these two processes can best be observed in patients recovering from stroke or ischemic injury.
Figure 7. Relationship between neurogenesis and angiogenesis in cancer. The interplay between angiogenesis and neurogenesis is due to signaling molecules, such as neurotrophins, neurotransmitters, and growth factors, which all act to stimulate both processes. These processes can also be stimulated by reactive oxygen species. Both the angiogenic and neurogenic signaling pathways also act to suppress the immune system. Apart from cancer, the interplay between these two processes can best be observed in patients recovering from stroke or ischemic injury.
Cancers 15 01805 g007
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

Dlamini, Z.; Khanyile, R.; Molefi, T.; Damane, B.P.; Bates, D.O.; Hull, R. Genomic Interplay between Neoneurogenesis and Neoangiogenesis in Carcinogenesis: Therapeutic Interventions. Cancers 2023, 15, 1805. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers15061805

AMA Style

Dlamini Z, Khanyile R, Molefi T, Damane BP, Bates DO, Hull R. Genomic Interplay between Neoneurogenesis and Neoangiogenesis in Carcinogenesis: Therapeutic Interventions. Cancers. 2023; 15(6):1805. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers15061805

Chicago/Turabian Style

Dlamini, Zodwa, Richard Khanyile, Thulo Molefi, Botle Precious Damane, David Owen Bates, and Rodney Hull. 2023. "Genomic Interplay between Neoneurogenesis and Neoangiogenesis in Carcinogenesis: Therapeutic Interventions" Cancers 15, no. 6: 1805. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers15061805

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

Article Metrics

Back to TopTop