Next Article in Journal
Left Ventricular Hypertrophy and Ventricular Tachyarrhythmia: The Role of Biomarkers
Next Article in Special Issue
Molecules Affecting Brain Development and Nervous System
Previous Article in Journal
Microbiota–Liver Diseases Interactions
Previous Article in Special Issue
SOCS7-Derived BC-Box Motif Peptide Mediated Cholinergic Differentiation of Human Adipose-Derived Mesenchymal Stem Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of SOCS and VHL Proteins in Neuronal Differentiation and Development

1
Department of Neurosurgery, School of Medicine, Yokohama City University, Yokohama 232-0024, Japan
2
Department of Neurosurgery, Asahi Hospital, Tokyo 121-0078, Japan
3
Department of Neurosurgery, St. Mariannna Medical University, Kawasaki 216-8511, Japan
4
Department of Neurosurgery, International University of Health and Welfare, Atami 413-0012, Japan
5
Nerve Care Clinic, Yokosuka 238-0012, Japan
6
Kochi Medical School Hospital, Nangoku 783-0043, Japan
7
Department of Electrical Engineering, University of California San Diego, San Diego, CA 92093, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(4), 3880; https://doi.org/10.3390/ijms24043880
Submission received: 22 December 2022 / Revised: 8 February 2023 / Accepted: 9 February 2023 / Published: 15 February 2023
(This article belongs to the Special Issue Molecules Affecting Brain Development and Nervous System)

Abstract

:
The basic helix–loop–helix factors play a central role in neuronal differentiation and nervous system development, which involve the Notch and signal transducer and activator of transcription (STAT)/small mother against decapentaplegic signaling pathways. Neural stem cells differentiate into three nervous system lineages, and the suppressor of cytokine signaling (SOCS) and von Hippel-Lindau (VHL) proteins are involved in this neuronal differentiation. The SOCS and VHL proteins both contain homologous structures comprising the BC-box motif. SOCSs recruit Elongin C, Elongin B, Cullin5(Cul5), and Rbx2, whereas VHL recruits Elongin C, Elongin B, Cul2, and Rbx1. SOCSs form SBC-Cul5/E3 complexes, and VHL forms a VBC-Cul2/E3 complex. These complexes degrade the target protein and suppress its downstream transduction pathway by acting as E3 ligases via the ubiquitin–proteasome system. The Janus kinase (JAK) is the main target protein of the E3 ligase SBC-Cul5, whereas hypoxia-inducible factor is the primary target protein of the E3 ligase VBC-Cul2; nonetheless, VBC-Cul2 also targets the JAK. SOCSs not only act on the ubiquitin–proteasome system but also act directly on JAKs to suppress the Janus kinase–signal transduction and activator of transcription (JAK-STAT) pathway. Both SOCS and VHL are expressed in the nervous system, predominantly in brain neurons in the embryonic stage. Both SOCS and VHL induce neuronal differentiation. SOCS is involved in differentiation into neurons, whereas VHL is involved in differentiation into neurons and oligodendrocytes; both proteins promote neurite outgrowth. It has also been suggested that the inactivation of these proteins may lead to the development of nervous system malignancies and that these proteins may function as tumor suppressors. The mechanism of action of SOCS and VHL involved in neuronal differentiation and nervous system development is thought to be mediated through the inhibition of downstream signaling pathways, JAK-STAT, and hypoxia-inducible factor–vascular endothelial growth factor pathways. In addition, because SOCS and VHL promote nerve regeneration, they are expected to be applied in neuronal regenerative medicine for traumatic brain injury and stroke.

1. Introduction

Since Cajal, a prominent 19th-century Spanish neuroanatomist, proposed that damaged central nerves cannot regenerate, it has long been believed that stem cells, the source of tissue regeneration, do not exist in the adult central nervous system. However, in 1965, newly generated neurons were observed in the hippocampus of adult rats [1]. Furthermore, in 1998, it was demonstrated that neural stem cells (NSCs) exist in the human subventricular zone (SVZ) and sub-granular zones of the hippocampal dentate gyrus [2], and it became clear that neurons are also produced in the adult human hippocampus.
To date, the understanding of the factors affecting neural differentiation and the development of the nervous system has developed rapidly via research on the differentiation mechanism of NSCs. Previous studies have identified a large number of cellular extrinsic factors and transcription factors that control NSC differentiation; this understanding facilitates our ability to induce the differentiation of NSCs into target cell types in vitro. For example, differentiation into neurons can be induced by the transcription of the pro-neural gene, neurogenin1 (Ngn1), followed by the induction of NeuroD gene expression [3]. The differentiation of NSCs into neurons is induced by promoting gene expression, and basic helix–loop–helix (bHLH)-type transcription factors are important for the differentiation of NSCs into oligodendrocytes and neurons [3,4]. The bHLH-type transcription factors, Olig1 and Olig2, whose expression is induced by Sonic Hedgehog (Shh) signaling, have been shown to regulate differentiation into oligodendrocytes and induce oligodendrocyte maturation [4]. bHLH-type transcription factors are inhibited by HES and ID family molecules [5,6]; furthermore, Notch and bone morphogenetic protein (BMP) signals induce the expression of these inhibitory bHLH factors [7,8], thereby promoting the neuronal differentiation of NSCs and oligodendrogenesis. It is involved in maintaining the undifferentiated state of NSCs and suppressing cell differentiation [7,8]. In contrast, interleukin-6 (IL-6) family cytokines (IL-6FCs) are known to induce the differentiation of neural connective cells into astrocytes [9]. The simultaneous stimulation of NSCs with IL-6FCs and BMP has been shown to synergistically induce differentiation into astrocytes [9]. Crosstalk in signaling pathways has also been revealed. In addition, IL-6FCs induce suppressor of cytokine signaling (SOCS) proteins and inhibit the Janus kinase–signal transduction and activator of transcription (JAK-STAT) pathway [10], thereby inhibiting the differentiation of NSCs into astrocytes. Overall, SOCS and von Hippel-Lindau (VHL) proteins work to suppress the JAK-STAT and/or hypoxia-inducible factor–vascular endothelial growth factor (HIF-VEGF) pathways [11,12], which are downstream of each protein, respectively. They then suppress the differentiation of NSCs into astrocytes and promote their differentiation into neurons and oligodendrocytes (Figure 1).
In this study, we describe the role of SOCS and VHL proteins in neuronal differentiation and development.

2. Structure of SOCS and VHL Proteins

SOCS (CIS and SOCS 1–7) and VHL form the Elongin B/C–Cul2/Cul5–SOCS-box protein (ECS) complex, a member of the ubiquitin ligase family that shares a Cullin-Rbx module. SOCS and VHL bind Cullin-Rbx via BC-box-binding Elongin C; however, VHL lacks the C-terminal sequence of the SOCS box (downstream of the BC box). In mammalian cells, SOCSs bind to Cul5-Rbx2; in contrast, VHL specifically interacts with endogenous Cul2-Rbx1. Therefore, the ECS complex has two distinct protein assemblies: one containing a subunit with a VHL box (composed of a BC box and a downstream Cul2 box) that interacts with Cul2–Rbx1 and the other containing a subunit with a SOCS box (composed of a BC box and a downstream Cul5 box). Domain exchange analyses demonstrated that the specificity of the interaction of VHL-box and SOCS-box proteins with the Cullin-Rbx module was conferred by Cul2 and Cul5 boxes, respectively. Additionally, the RNAi-mediated knockdown of Cul2–Rbx1 inhibited the VHL-mediated degradation of HIF-2α, whereas Cul5–Rbx2 knockdown did not affect the function of the Cul2–Rbx1 and Cul5–Rbx2 modules [13] (Figure 2).

3. Characteristics of SOCS Proteins and Their Roles in Neuronal Differentiation and Nervous System Development

3.1. Features and Functions of SOCS Proteins

The discovery of the SOCS family began in 1995 with the isolation of a novel cytokine-inducible Src homology 2 (SH2)-containing protein (CIS) as an early gene induced by IL-2, IL-3, and erythropoietin (EPO) [13]. Subsequently, an SH2 domain–containing protein that inhibits IL-6-induced macrophage differentiation was identified in 1997 and named SOCS1 [14,15,16]. Furthermore, database searches of similar amino acid sequences predicted from SOCS1 revealed that at least 20 proteins from mice and humans share sequence homology within a 40-residue C-terminal motif called the SOCS box. Proteins containing SOCS boxes were grouped into subfamilies based on the domains contained in the central region, and family member proteins containing central SH2 domains were named SOCSs (CIS and SOCS1–7). Each SOCS has three distinct domains: a least-conserved N-terminal domain, a conserved central SH2 domain, and a highly conserved C-terminal SOCS-box domain. The length of the N-terminal domain varies among the members; SOCS1–3 and CIS contain shorter N-terminal domains than in SOCS4–7 [17]. The SOCS-box motif recruits an E3 ubiquitin ligase complex consisting of Elongin B/C, Rbx2, and Cullin5. This complex forms an E3 ubiquitin ligase that ubiquitin-tags target proteins, such as JAKs and cytokine receptors, and degrades them in the proteasome [18,19]. SOCSs regulate signaling by linking substrates to the ubiquitination machinery through the SOCS box. SOCS expression, induced by cytokine stimulation, not only interferes with cytokine signaling in a negative feedback loop but also interferes with other cytokines downstream in a response known as “crosstalk” [20,21]. Currently, various molecules, including numerous cytokines, such as interferons (IFNs), growth factors, and hormone factors, are known to activate the JAK-STAT pathway; in contrast, the JAK-STAT pathway is inhibited by SOCS [22] (Figure 3).
Among the SOCS family members, CIS, SOCS1, and SOCS3 inhibit the JAK-STAT signaling pathway; however, their modes of action are slightly different. SOCS1 directly interacts with JAK1, JAK2, and tyrosine kinase 2 (Tyk2) and inhibits their phosphorylation and catalytic activity, thereby downregulating the JAK-dependent phosphorylation of receptors and the signal transducer and activator of transcription (STATs) [19,20,23].
Structurally, the SH2 domain and N-terminal region of SOCS1 are essential for inhibition of JAK activity, whereas the C-terminal SOCS-box has been shown to be dispensable [23,24,25].
CIS expression is induced by EPO [24], growth hormone (GH) [25,26,27,28,29], IL-2 [30,31,32,33,34], IL-3 [31,32], and prolactin (PRL) [32,33]. SOCS1 expression also inhibits IFN signaling by interacting with the IFN-α/β receptor subunit 1 and IFN-γ receptor subunits, which in turn inhibit IFN activation via STAT3 [34]. Additionally, SOCS1 promotes the degradation of p65 in the NF-κB signaling pathway and reduces the kinase apoptotic signal that regulates kinase 1 upstream in the JNK and p38 signaling pathways; consequently, SOCS1-mediated degradation inhibits GH [35,36,37], IFN [29,30,31], IL-4 [32,33,34], IL-6 [29,32], thrombopoietin (TPO) [29], PRL [38]. SOCS2 expression is then also induced by STAT5-activating cytokines such as ciliary neurotrophic factor (CNTF) [39], IFNα [40], IFNγ [18,41], leukemia inhibitory factor (LIF) [18], IL-1β [41], IL-4 [18], IL-6 [41], IL-15 [42], and insulin (INS) [43]. In contrast, SOCS2 downregulates the expression of the cytokines GH [44], PRL [45], LIF [45], IL-2, IL-3 [46], IL-6 [47], epidermal growth factor (EGF) [48], and insulin-like growth factor (IGF)-1 via a negative feedback loop [49].
SOCS3 has been demonstrated to be induced by the cytokines IL-1β [50], IL-2 [51], IL-3 [52], IL-4 [32], IL-6 [53], IL-9 [54], IL-10 [55], IL-11 [56], IL-13 [18], IL-22 [57], IFN-γ [31], IFNα [31], EPO [38], LIF [58], PRL [34], GH [58], leptin [59], G-CSF [18], GM-CSF [60], CNTF [39], TPO [61], TNFα [62], CT-1 [63], and oncostatin M signaling (OSM) [64]. It is also induced by several growth factors, including EGF [65], platelet-derived growth factor (PDGF) [65], thyroid-stimulating hormone (TSH) [66], insulin [43], and basic fibroblast growth factor (bFGF) [67]. SOCS3 has been demonstrated to play a regulatory role in the downstream signaling of a wide range of cytokine receptors, including those for IL-2 [42], IL-6 [47], IL-9 [54], IL-11 [56], IL12 [68], IL-23 [69], IL-27 [70], IFNα/β [31], IFNγ [31], G-CSF [71], EPO [34], PRL [34], GH [72], LIF [58], leptin [59], CNTF [61], IL-1β [73], OSM [65], and CT-1 [64], as well as IGF-1 [74], INS [75], CD28 [27] and calcineurin [76]. Conversely, SOCS3 inhibits the nuclear factor κB (NF-κB) signaling pathway, antagonizes cyclic AMP (cAMP)-mediated signaling, and promotes signaling via the microtubule affinity-regulating kinase (MAPK) pathway [77].
SOCS4 has been shown to be induced exclusively by EGF [78] and has been demonstrated to regulate EGFR signaling in vitro [79]. This induction is mediated by SOCS4 docking on phosphotyrosine residues on activated EGFR and the subsequent targeting of the receptor for proteasomal degradation by the recruitment of E3 ubiquitin ligase activity [79,80]. Similar to STAT3, SOCS4 binds to EGFR phosphotyrosine with a high affinity. In addition, SOCS4 regulates primordial follicle activation initiated by LIF activation of the JAK1-STAT3 pathway and interacts with several proteins involved in follicle development [81].
SOCS5 expression has been shown to be induced by EGF in vitro [78]. SOCS5 regulates both receptor tyrosine kinase (RTK) and cytokine receptor signaling and negatively regulates EGFR in vitro [80,81]. In addition, SOCS5 downregulates the receptors of IL-6, LIF [82], and IL-4 signaling [78,79]. SOCS5 has also been hypothesized to regulate signaling by initiating IL-4 signaling. SOCS5 has also been implicated in T helper (Th) cell differentiation, particularly in the balance between Th1 and Th2 cells. SOCS5 inhibits activation of STAT6 by the IL-4 receptor and typically induces T-cell differentiation toward a Th2 fate [83,84]. Additionally, SOCS5 transgenic mice show increased IL-2 and IFN-γ expression [85]. SOCS6 is induced by INS [85,86], IGF-2 [87], IGF-1 [87], FMS-like tyrosine kinase 3 (FLT3) [88], and stem cell factor (SCF) [89]. SOCS5 also negatively regulates T-cell antigen receptor (TCR) signaling [90].
SOCS6 exerts its regulatory effects predominantly through the ubiquitination and degradation of target proteins [41]. SOCS6 also interacts with another E3 ligase component, heme-oxidized IRP2 ubiquitin ligase-1 (HOIL-1) [89]. Further, the SOCS6 N-terminal domain promotes localization to the nucleus, where it negatively regulates STAT3. The overexpression of SOCS6 [90] results in the repression of TCR-dependent IL-2 promotion and activation of MAPK pathway factors, such as ERK1/2 and p38 [34]. SOCS6 also binds to FLT3 and negatively regulates its corresponding signaling [88]. Furthermore, SOCS6 has been shown to inhibit the downstream pathways of INS and IGF-1 receptors [86]. Specifically, SOCS6 interacts with PIM3, a protein upregulated in β-cells in response to glucose stimulation.
SOCS7 is induced by the cytokines GH, PRL [41], EGF [91], INS, and IGF-1 [41] and regulates signaling via GH, PRL, leptin [91], and INS [92]. SOCS7 also inhibits the nuclear transport of the following adapter proteins: non-catalytic region of tyrosine kinase (NCK) [93], insulin receptor substrate (IRS)-1 [91], IRS-2 [92], IRS-4 [92], p85 phosphatidylinositol 3-kinase (PI3K) [92], and growth factor receptor-bound protein 2 (GRB2) [93]. SOCS7 regulates signaling through the recruitment of E3 ubiquitin ligase activity and subsequent proteasome targeting of associated proteins [92]. SOCS7 interacts with INS receptors and their adapter proteins, playing an active role in insulin signaling [92,93]. In addition, associations have been observed between SOCS7 haplotypes and various metabolic traits, such as obesity, insulin resistance, and lipid metabolism [94].

3.2. SOCS Gene and Protein Expression in the Nervous System

Members of the SOCS family may play important roles in the nervous system, primarily owing to their regulatory action on the JAK-STAT pathway, which is involved in nervous system development and differentiation. The expression of SOCS1, SOCS2, and SOCS3 in the development of adult nervous systems was investigated using northern analysis and in situ hybridization [95]. In the corresponding northern analysis, all three genes were expressed in the brain, with SOCS2 being the most highly expressed; maximal expression was observed from embryonic day 14 to postnatal day 8 and decreased thereafter. In situ hybridization analysis revealed that the expression patterns of SOCS1 and SOCS3 were low and widespread, whereas SOCS2 was only expressed in neurons; the corresponding expression of SOCS2 was induced during neuronal differentiation [95]. SOCS4 is widely expressed in the brain, with a particularly high expression in the olfactory nerve. SOCS5 is not only highly expressed in the placenta but also in the brain [21,79,96,97]. In contrast, in the nervous system, SOCS6 and SOCS7 are highly expressed in the retina and throughout the brain, respectively [92,93].

3.3. Role of SOCS Proteins in the Molecular Mechanisms of Neuronal Differentiation and Nervous System Development

SOCSs are involved in the differentiation of various cells. Regarding T-cell differentiation, SOCS1 in Th cells is induced by IL-6 and promotes Th17 differentiation [98]. In contrast, SOCS3 promotes the differentiation of myeloid cells into lung tissue CD8+ T lymphocytes via Notch1 upregulation [99]. SOCSs also play important roles in neuronal differentiation and regeneration, neurogenesis, and nervous system development. SOCS1 regulates IFN-γ-mediated survival of sensory neurons [100] and NSC proliferation and differentiation into neurons [101,102].
SOCS2 is a negative regulator of GH signaling [103,104,105]. In contrast, SOCS2 was shown to promote neurite outgrowth, regulate neuronal morphology, induce neurogenesis, and inhibit NSC astrogliogenesis [103,106]. SOCS3 associates with IGF receptors and inhibits the JAK-STAT pathway [107]. Furthermore, SOCS3 induces neuronal differentiation and promotes the neuronal survival of PC12 cells [108]. SOCS6 also promotes the neuronal differentiation of NSCs [87]. Additionally, we showed that the BC-box motif in SOCS6 promotes neuronal differentiation into GABAergic neurons [109].
The neurogenic cytokine family, which includes IL-6, LIF, and CNTF, also regulates the JAK-STAT pathway. The neurogenic cytokine family proteins are activated to alter NSC self-renewal and progenitor cell division and differentiation; furthermore, these proteins induce SOCS expression to exert negative feedback on the JAK-STAT pathway. Experiments with NSCs isolated from the adult mouse SVZ showed that both cytokines activated the JAK-STAT pathway and stimulated NSC self-renewal when LIF or CNTF was administered in the culture medium. Thus, SOCS promotes the growth and development of NSC neurospheres. Consequently, LIF treatment for acute brain injury has been observed to promote SVZ regeneration, possibly by increasing the number of NSCs [110].
The classical neurotrophins nerve growth factor (NGF), brain-derived neurotrophic factor, neurotensin (NT)-3, and NT-4 regulate multiple aspects of neuronal differentiation, survival, and growth. To exert these effects, these factors specifically bind to receptors of the tropomyosin-related kinase (Trk) receptor tyrosine kinase family, namely TrkA, TrkB, and TrkC. They are internalized and localized in various cellular compartments where signaling occurs. Conversely, among the SOCS family members, SOCS2, which is also a regulator of NGF signaling, alters the subcellular localization and downstream signaling of TrkA and plays a role in neuronal differentiation and neurite outgrowth, similar to NGF [111].
The expression of SOCS1 and SOCS3 in glioblastoma (GBM) cells is decreased. In addition, the JAK-STAT3 signaling pathway is activated in human gliomas, leading to poor prognosis [112]. SOCS1 and SOCS3 may also function as epigenetic regulators of GBM hypermethylation, associated with poor prognosis in GBM [113]. NF-κB-driven SOCS expression functions as a negative regulator of the JAK-STAT signaling pathway. In GBM, E3 ubiquitin ligases are involved in regulating cellular functions, such as RTK survival signals. SOCSs negatively regulate RTK signaling, and kinase overexpression or mutation is associated with glioma malignancy [114].

4. Characteristics of VHL Proteins and Their Roles in Neuronal Differentiation and Nervous System Development

4.1. Features and Functions of VHL Proteins

VHL is the product of the VHL tumor suppressor gene, the causative gene of VHL disease. VHL disease is an autosomal dominant hereditary multiple neoplastic disease that causes hemangioblastoma, retinal hemangioma, renal cell carcinoma, pheochromocytoma, and pancreatic, renal, and epididymal cysts. This disease was named after Eugene von Hippel, who reported familial retinal angioma in 1904, and Arvid Lindau, who reported a case of retinal hemangioma and cerebellar hemangioblastoma in 1926 [115].
VHL disease is caused by mutations in the VHL tumor suppressor gene. VHL was isolated and identified from the 3p25 region using the positional cloning method described by Zbar et al. [116]. VHL consists of 639 base pairs and three exons. The genetic diagnosis of patients with this disease using this gene revealed mutations concentrated in the binding site of the binding protein. In approximately 20% of cases, the entire VHL locus is deleted, which can be detected by quantitative Southern blot analysis or fluorescence in situ hybridization [115,116]. Approximately 40% of the VHL mutations are frameshift or nonsense mutations, and the remaining 40% are missense mutations [116,117].
VHL consists of three exons and is located in a region of approximately 13,000 bp on 3p25.3 in the human genome. However, translation is initiated by two methionine residues at amino acids 1 and 54; therefore, VHL proteins of 213 and 160 amino acids (approximately 30 and 19 kDa in size, respectively) are produced, both of which have tumor-suppressing functions [115].
VHLs shuttle between the nucleus and cytoplasm, and, at steady state, the majority of the protein resides in the cytoplasm [118,119,120,121,122,123,124,125,126]. Some VHL proteins are also found in the mitochondria and are associated with the endoplasmic reticulum [127,128]. The primary sequence of VHL contains two functional subdomains, called the alpha and beta domains (Figure 2) [129]. The alpha domain, corresponding to residues 155–192, consists of three alpha helices and forms the core of a protein complex containing Elongin C, Elongin B, Cul2, and Rbx1 (also known as Roc1) [130,131,132,133,134]. This complex possesses ubiquitin ligase activity, which leads to proteasomal degradation of the target protein [135,136].
The most well-assessed function of VHL is its function as an E3 ubiquitin ligase complex that regulates the degradation of the transcription factor HIF. The VHL protein consists of two structural functional domains, α and β. The α-domain binds to Elongin C, Elongin B, Cullin2(Cullin2), and Rbx1 to form an E3 ubiquitin ligase complex (VBC-Cul2/E3 complex) [127,133,137]. VHL binds to the target protein at the β-domain; however, one of the ubiquitinated target proteins is HIF-1α, which undergoes post-translational modification (hydroxylation of proline residues). HIF forms a heterocomplex of two molecules: HIF-1α and HIF-1β. HIF-1α binds to the cofactor CBP/p300 and has functional activity as a transcription factor. Additionally, HIF-1α is post-translationally modified by HIF prolyl hydroxylase under normoxic conditions by hydroxylating proline residues (amino acids 402 and 564 in HIF-1α, and 405 and 531 in HIF-2α). Hydroxylated HIF-α (post-translationally modified) is polyubiquitinated by the VBC-Cul2/E3 complex and then rapidly degraded by the 26S proteasome [138,139]. Conversely, under hypoxic conditions, the ubiquitination and degradation of HIF-1α are suppressed; HIF-1α translocates to the nucleus, binds to HIF-1β, binds to the hypoxia response element in the gene promoter, and initiates the transcription of various genes. [140] (Figure 4).

4.2. Expression of the VHL Gene and Protein in the Nervous System

With regard to VHL expression, strong expression of VHL mRNA during human embryogenesis was found in all three germ layers; in particular, its expression was observed in the central nervous system, kidney, testis, and lung via in situ hybridization analysis at 4, 6, and 10 weeks post conception. Moreover, studies of the expression distribution pattern of VHL have shown that VHL is widely expressed in normal human tissues and that high levels of VHL are found in neural tissue. In particular, VHL expression was observed in Purkinje cells and Golgi type II cells; additionally, regional expression was observed in the cerebellum, dentate nucleus, pontine nucleus, inferior olivary nucleus of the medulla oblongata, sympathetic ganglion, mesomysium, and colonic submucosal plexus. In target organs of VHL disease, other than the nervous system, high VHL expression was observed in the renal tubular system, pancreatic exocrine gland, adrenal cortex, and liver parenchyma [141]. In addition, the expression of the VHL was observed to be limited to the adult and fetal brain, kidney, and adult prostate. Nerve cells, including cerebellar Purkinje cells, in adult and fetal brains were positive for VHL gene expression [142]. In contrast to prior studies, VHL was observed to be widely expressed in normal human tissues. Cellular distribution of the protein was confined to the cytoplasm of specific cell types. High levels of the protein were observed in neural tissue, especially in Purkinje cells and Golgi type II cells, the dentate nucleus of the cerebellum and pontine nuclei, inferior olivary nucleus of the medulla oblongata, orthosympathetic ganglia, and myenteric and submucous plexus of the colon [122,143]. In addition, strong expression of VHL was found in neural progenitor cells that differentiated into neurons on day 14 of culture [144] (Figure 5).

4.3. Role of VHL Protein in the Molecular Mechanisms of Neuronal Differentiation and Nervous System Development

We showed that VHL expression in rat forebrain-derived NSCs is predominant in the cytoplasm of cells expressing the neuronal marker microtubule-associated proteins 2 (MAP2) and that the forced expression of VHL rapidly induces differentiation into neurons [144]. In addition, the electrophysiological analysis revealed that VHL-introduced NSCs had an average maximum current of ≥4000 pA; additionally, the voltage-dependent sodium current of these cells was determined to be >4000 pA. These cells showed more than three times the average maximum current of cells that had naturally differentiated into cells and exhibited properties of electro-physiologically mature neurons, suggesting the possibility of their roles as functional neurons in vitro [145]. Furthermore, more than 50% of VHL transgenic NSCs transplanted into Parkinson’s disease rat models not only differentiated into tyrosine hydroxylase (TH)-positive dopamine-producing cells but also showed a marked reduction in apomorphine-induced turnover in behavioral analysis. Approximately 30% of the model rats showed no induced rotation [146]. This finding indicated that VHL-introduced NSCs function as dopamine-producing neurons in the transplanted rat brain. Therefore, neural transplantation therapy for neurological diseases, such as Parkinson’s disease, using NSCs as donors is of great importance, with VHL expression potentially contributing to the understanding of these treatments.
The VHL forms a complex (VBC-Cul2/E3 complex) with Elongin B and C, Cullin2, and Rbx1, which acts as an E3 ligase. After the ubiquitination of JAK2, its degradation by proteasome 26S, followed by the suppression of downstream STAT expression, may affect bHLH-type transcription factors, in turn affecting the Notch signaling and STAT/Smad pathways [13,14,147,148]. VBC-Cul2/E3 complexes ubiquitinate HIF-1α for proteasomal 26S degradation, followed by the activation of downstream factors of HIF-1α. The suppression of HIF-1α expression is thought to induce neuronal rather than glial differentiation [149]. The VHL forms a VBC-Cul2 complex with its binding proteins, Elongin B/C and Cul2, and degrades HIF. However, normal oxygen tension is required for the normal functioning of VHL. Under hypoxia, VHL does not function normally, and therefore, HIF is not degraded; then, hypoxia-induced VEGF, EPO, and other factors are induced [150] (Figure 4). Therefore, in our study, we placed NSCs under hypoxic conditions and investigated their differentiation when VHL did not function normally. Consequently, the percentage of the neuronal marker MAP-positive cells decreased under hypoxic conditions, but the percentage of the astroglial marker GFAP-positive cells increased. VHL protein dysfunction due to hypoxia may result in the inhibition of neuronal differentiation [151]. Since VHL does not function under hypoxia, HIF is not degraded, and downstream transcription factors are induced, suggesting that HIF may be involved in the differentiation of NSCs into glia. As parenchymal tumors of the central nervous system, such as gliomas, can arise from NSCs, this VHL-HIF transcriptional mechanism is crucial in the development of parenchymal tumors of the central nervous system. [152,153]
Furthermore, we identified that the domain of VHL involved in neuronal differentiation is the BC-box motif. Interestingly, it was found that neural differentiation occurs when the BC-box motif in the VHL protein is introduced into NSCs and other somatic stem cells (skin-derived stem cells, bone marrow-derived stem cells, adipose-derived stem cells, and human epidermal stem cells) [145,154,155,156,157,158]. In addition, in the SOCS family proteins that have a BC-box motif structure homologous to the VHL protein, most of the BC-box motifs exhibited neuronal differentiation activity [148].
It has also recently been reported that VHL interacts with Daam2; specifically, their mutual antagonism regulates differentiation in oligodendrocyte development. Proteomic analysis of the Daam2–VHL complex using a conditional gene knockout mouse model revealed that the Nedd4 complex ubiquitinates amino acid K63 and stabilizes VHL; this process is required for myelination. Furthermore, studies in mouse demyelination models and white matter lesions in patients with multiple sclerosis have demonstrated the role of the Nedd4-VHL pathway in remyelination; in particular, Nedd4 is required for differentiation and myelination of oligodendrocytes. In addition, it has been established that VHL-HIF signaling plays a key role in the tumorigenesis of a wide range of malignancies. The Daam2–VHL relationship was first discovered in gliomas, with Nedd4 being a key upstream regulator of VHL. This discovery may have broad implications, including both CNS and non-CNS malignancies. It is important to clarify whether Nedd4-VHL-HIF signaling is involved in the development of renal carcinoma and hemangioblastoma, which commonly occur in VHL disease [159].
Recently, attention has been focused on the role of HIF-1α in NSCs during nerve regeneration after traumatic brain injury and stroke; furthermore, VHL, upstream of HIF-1α, is thought to play an important role in this process. Endogenous NSCs and neural progenitor cells (NSPCs) have been shown to promote repair after brain injury and stroke. Therefore, these cells are therapeutic targets for promoting cell replacement therapy and neuroplasticity. The mechanisms supporting NSPC survival and recruitment of damaged cells in the ischemic brain have highlighted a novel role for HIF-1α in NSPC function. HIF-1α mediates adaptive cellular responses to hypoxia through the direct transcriptional regulation of cell metabolism and angiogenesis; however, recent studies have suggested that it may be mediated by regulation of the Notch and Wnt/β-catenin signaling pathways [152].
A novel role of HIF-1α in stem cell differentiation has also been demonstrated. HIF-1α functions as a key mediator of NSPC function under normal conditions and in stroke, playing a central role in regulating NSPC responses to hypoxia, metabolism, and the maintenance of the vascular environment of the NSC niche. The role of VHL proteins upstream of HIF-1α has also attracted attention. Focal cerebral ischemia stimulates the proliferation and migration of SVZ-derived progenitor cells into the lesion, suggesting that NSPCs in the SVZ recruit new oligodendrocyte progenitor cells, astrocytes, and neural progenitor cells to the peri-infarct region. In addition, HIF-1α not only maintains the vascular niche environment and promotes angiogenesis through transcriptional regulation of VEGF but also is involved in determining the pluripotency and differentiation of NSPCs. As HIF-1α plays an important role in such processes, VHL, upstream of HIF-1α, is thought to play a similarly important role [160].

5. Conclusions

bHLH factors play a central role in mammalian nervous system development and differentiation from NSCs to nervous system cells. The Notch and STAT/Smad pathways are thought to be involved in this process; additionally, the STAT transduction pathway is regulated by SOCS and VHL proteins. NSCs differentiate into three tumor cell lineages: neurons, astrocytes, and oligodendrocytes. SOCS is involved in neuronal differentiation, and VHL is involved in differentiation into neurons and oligodendrocytes; however, both proteins function to suppress astrocyte differentiation. SOCS and VHL proteins contain homologous structures comprising a BC-box motif, called SOCS box and VHL box, respectively. SOCS proteins specifically bind to Elongin B/C-Cul5-Rbx2 to form an SBC-Cul5/E3 complex, whereas VHL specifically binds to Elongin B/C-Cul2-Rbx1 to form a VBC-Cul2/E3 complex. Both complexes act as E3 ligases that target proteins via the ubiquitin–proteasome system, ultimately resulting in the degradation and inhibition of downstream transduction pathways. We have shown that SBS-Cul5/E3 and VBC-Cul2/E3 recruit JAK and HIF as their primary target proteins, respectively; however, JAK is also a target protein of VBC-Cul2. Additionally, SOCSs not only act on the ubiquitin–proteasome system but also act directly on JAKs to suppress the JAK-STAT pathway. SOCS and VHL proteins are expressed in the nervous system, with predominant expression in embryonic brain neurons. Both SOCS and VHL proteins promote neurite outgrowth and induce neuronal differentiation. SOCS proteins are involved in differentiation into neurons, and VHL proteins are involved in differentiation into neurons and oligodendrocytes. In addition, SOCS and VHL proteins function as tumor suppressors; consequently, it has been suggested that the inactivation of these proteins may lead to the development of nervous system malignancies. The mechanism of action of SOCS and VHL proteins involved in neuronal differentiation and development of the nervous system is thought to be mediated via the inhibition of downstream signaling pathways (JAK-STAT and HIF-VEGF pathways). In addition, because SOCS and VHL proteins promote nerve regeneration, they are expected to be applied in nerve-regenerative medicine for traumatic brain injury and stroke.

Author Contributions

Conception and design, Data analysis and interpretation, Manuscript writing: H.K. Conception and design, Supervising writing manuscript: T.S. and H.-S.U. Conception and design, Data analysis and interpretation: K.N., A.K. and H.M. Collection and assembly of data, Data analysis and interpretation: T.Y. and A.K. Conception and design, Data analysis and interpretation: T.Y. and H.M. Data curation, Formal analysis, Software: S.M. Project administration, Software: H.K., H.M., T.S. and H.-S.U. Collection and assembly of data: H.K., S.M., K.N., T.Y., A.K. and H.M. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by grants from the Basic Research (C) No. 21K09134; Ministry of Education, Culture, Sports, Science, and Technology of Japan.

Data Availability Statement

The data presented in this study are openly available only in this text.

Conflicts of Interest

The authors have no conflicting financial interest.

References

  1. Altman, J.; Das, G.D. Autoradiographic and histological evidence of postnatal hippocampal neurogenesis in rats. J. Comp. Neurol. 1965, 124, 319–335. [Google Scholar] [CrossRef] [PubMed]
  2. Pincus, D.W.; Keyoung, H.M.; Harrison-Restelli, C.; Goodman, R.R.; Fraser, R.A.; Edgar, M.; Sakakibara, S.; Okano, H.; Nedergaard, M.; Goldman, S.A. Fibroblast growth factor-2/brain-derived neurotrophic factor-associated maturation of new neurons generated from adult human subependymal cells. Ann. Neurol. 1998, 43, 576–585. [Google Scholar] [CrossRef] [PubMed]
  3. Sun, Y.; Nadal-Vicens, M.; Misono, S. Neurogenin promotes neurogenesis and inhibits glial differentiation by independent mechanism. Cell 2001, 104, 365–376. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Seo, S.; Lim, J.W.; Yellajoshyula, D.; Chang, L.W.; Kroll, K.L. Neurogenin and NeuroD direct transcriptional targets and their regulatory enhancers. EMBO J. 2007, 26, 5093–5108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Lu, Q.; Yuk, D.; Alberta, J.A. Sonic hedgehog-regulated oligodendrocyte lineage genes encoding bHLH proteins in the mammalian central nervous system. Neuron 2000, 25, 317–329. [Google Scholar] [CrossRef] [Green Version]
  6. Bai, G.; Sheng, N.; Xie, Z.; Bian, W.; Yokota, Y.; Benezra, R.; Kageyama, R.; Guillemot, F.; Jing, N. Id sustains Hes1 expression to inhibit precocious neurogenesis by releasing negative autoregulation of Hes1. Dev. Cell 2007, 13, 283–297. [Google Scholar] [CrossRef] [Green Version]
  7. Morrison, S.J.; Perez, S.E.; Qiao, Z. Transient Notch activation initiates an irreversible switch from neurogenesis to gliogenesis by neural crest stem cells. Cell 2000, 101, 499–510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Nakashima, K.; Takizawa, T.; Ochiai, W.; Yanagisawa, M.; Hisatsune, T.; Nakafuku, M.; Miyazono, K.; Kishimoto, T.; Kageyama, R.; Taga, T. BMP2-mediated alteration in the development pathway of astrocytogenesis. Proc. Natl. Acad. Sci. USA 2001, 98, 5868–5873. [Google Scholar] [CrossRef] [Green Version]
  9. Taga, T.; Fukuda, S. Role of IL-6 in the neural stem cell differentiation. Clin. Rev. Allergy Immunol. 2005, 28, 249–256. [Google Scholar] [CrossRef]
  10. Yanagisawa, M.; Nakashima, K.; Takizawa, T.; Ochiai, W.; Arakawa, H.; Taga, T. Signaling crosstalk underlying synergistic induction of astrocyte differentiation by BMPs and IL-6 family of cytokines. FEBS Lett. 2001, 489, 139–144. [Google Scholar] [CrossRef]
  11. Heinrich, P.C.; Behrmann, I.; Haan, S.; Hermanns, H.M.; Müller-Newen, G.; Schaper, F. Principles of interleukin (IL)-6-type cytokine signalling and its regulation. Biochem. J. 2003, 374 Pt 1, 1–20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Cooney, R.N. Suppressors of cytokine signaling (SOCS): Inhibitors of the JAK/STAT pathway. Shock 2002, 17, 83–90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Kanno, H.; Sato, H.; Yokoyama, T.; Yoshizumi, T.; Yamada, S. The VHL tumor suppressor protein regulates tumorigenicity of U87-derived glioma stem-like cells by inhibiting the JAK/STAT signaling pathway. Int. J. Oncol. 2013, 42, 881–886. [Google Scholar] [CrossRef] [Green Version]
  14. Kanno, H.; Yoshizumi, T.; Shinonaga, M.; Kubo, A.; Murata, H.; Yao, M. Role of VHL-JAK-STAT signaling pathway in central nervous system hemangioblastoma associated with von Hippel-Lindau disease. J. Neurooncol. 2020, 148, 29–38. [Google Scholar] [CrossRef]
  15. Kim, H.; Shim, B.Y.; Lee, S.J.; Lee, J.Y.; Lee, H.J.; Kim, I.H. Loss of von Hippel-Lindau (VHL) tumor suppressor gene function: VHL-HIF pathway and advances in treatments for metastatic renal cell carcinoma (RCC). Int. J. Mol. Sci. 2021, 22, 9795. [Google Scholar] [CrossRef]
  16. Kamura, T.; Maenaka, K.; Kotoshiba, S.; Matsumoto, M.; Kohda, D.; Conaway, R.C.; Conaway, J.W.; Nakayama, K.I. VHL-box and SOCS-box domains determine binding specificity for Cul2-Rbx1 and Cul5-Rbx2 modules of ubiquitin ligases. Genes Dev. 2004, 18, 3055–3065. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Yoshimura, A.; Ohkubo, T.; Kiguchi, T.; Jenkins, N.A.; Gilbert, D.J.; Copeland, N.G.; Hara, T.; Miyajima, A. A novel cytokine-inducible gene CIS encodes an SH2-containing protein that binds to tyrosine-phosphorylated interleukin 3 and erythropoietin receptors. EMBO J. 1995, 14, 2816–2826. [Google Scholar] [CrossRef]
  18. Starr, R.; Willson, T.A.; Viney, E.M.; Murray, L.J.; Rayner, J.R.; Jenkins, B.J.; Gonda, T.J.; Alexander, W.S.; Metcalf, D.; Nicola, N.A.; et al. A family of cytokine-inducible inhibitors of signalling. Nature 1997, 26, 917–921. [Google Scholar] [CrossRef]
  19. Endo, T.A.; Masuhara, M.; Yokouchi, M.; Suzuki, R.; Sakamoto, H.; Mitsui, K.; Matsumoto, A.; Tanimura, S.; Ohtsubo, M.; Misawa, H.; et al. A new protein containing an SH2 domain that inhibits JAK kinase. Nature 1997, 26, 921–924. [Google Scholar] [CrossRef]
  20. Naka, T.; Narazaki, M.; Hirata, M.; Matsumoto, T.; Minamoto, S.; Aono, A.; Nishimoto, N.; Kajita, T.; Taga, T.; Yoshizaki, K.; et al. Structure and function of a new STAT-induced STAT inhibitor. Nature 1997, 387, 924–929. [Google Scholar] [CrossRef]
  21. Hilton, D.J.; Richardson, R.T.; Alexander, W.S.; Viney, E.M.; Willson, T.A.; Sprigg, N.S.; Starr, R.; Nicholson, S.E.; Metcalf, D.; Nicola, N.A. Twenty proteins containing a C-terminal SOCS box form five structural classes. Proc. Natl. Acad. Sci. USA 1998, 95, 114–119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Kamura, T.; Sato, S.; Haque, D.; Liu, L.; Kaelin, W.G., Jr.; Conaway, R.C.; Conaway, J.W. The Elongin BC complex interacts with the conserved SOCS-box motif present in members of the SOCS, ras, WD-40 repeat, and ankyrin repeat families. Genes Dev. 1998, 12, 3872–3881. [Google Scholar] [CrossRef] [Green Version]
  23. Zhang, J.G.; Farley, A.; Nicholson, S.E.; Willson, T.A.; Zugaro, L.M.; Simpson, R.J.; Moritz, R.L.; Cary, D.; Richardson, R.; Hausmann, G.; et al. The conserved SOCS box motif in suppressors of cytokine signaling binds to elongins B and C and may couple bound proteins to proteasomal degradation. Proc. Natl. Acad. Sci. USA 1999, 96, 2071–2076. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Sasi, W.; Sharma, A.K.; Mokbel, K. The role of suppressors of cytokine signalling in human neoplasms. Mol. Biol. Int. 2014, 2014, 630797. [Google Scholar] [CrossRef]
  25. Piessevaux, J.; Lavens, D.; Montoye, T.; Wauman, J.; Catteeuw, D.; Vandekerckhove, J.; Belsham, D.; Peelman, F.; Tavernier, J. Functional cross-modulation between SOCS proteins can stimulate cytokine signaling. J. Biol. Chem. 2006, 281, 32953–32966. [Google Scholar] [CrossRef] [Green Version]
  26. Matsumoto, A.; Masuhara, M.; Mitsui, K.; Yokouchi, M.; Ohtsubo, M.; Misawa, H.; Miyajima, A.; Yoshimura, A. CIS, a cytokine inducible SH2 protein, is a target of the JAK-STAT5 pathway and modulates STAT5 activation. Blood 1997, 89, 3148–3154. [Google Scholar] [CrossRef] [Green Version]
  27. Matsumoto, A.; Seki, Y.; Kubo, M.; Ohtsuka, S.; Suzuki, A.; Hayashi, I.; Tsuji, K.; Nakahata, T.; Okabe, M.; Yamada, S.; et al. Suppression of STAT5 functions in liver, mammary glands, and T cells in cytokine inducible SH2-containing protein 1 transgenic mice. Mol. Cell. Biol. 1999, 19, 6396–6407. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Ram, P.A.; Waxman, D.J. Role of the cytokine-inducible SH2 protein CIS in desensitization of STAT5b signaling by continuous growth hormone. J. Biol. Chem. 2000, 275, 39487–39496. [Google Scholar] [CrossRef] [Green Version]
  29. Aman, M.J.; Migone, T.S.; Sasaki, A.; Ascherman, D.P.; Zhu, M.; Soldaini, E.; Imada, K.; Miyajima, A.; Yoshimura, A.; Leonard, W.J. CIS associates with the interleukin-2 receptor beta chain and inhibits interleukin-2- dependent signaling. J. Biol. Chem. 1999, 274, 30266–30272. [Google Scholar] [CrossRef] [Green Version]
  30. Ram, P.A.; Waxman, D.J. SOCS/CIS protein inhibition of growth hormone-stimulated STAT5 signaling by multiple mechanisms. J. Biol. Chem. 1999, 274, 35553–35561. [Google Scholar] [CrossRef]
  31. Song, M.M.; Shuai, K. The suppressor of cytokine signaling (SOCS) 1 and SOCS3 but not SOCS2 proteins inhibit interferon mediated antiviral and antiproliferative activities. J. Biol. Chem. 1998, 273, 35056–35062. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Losman, J.A.; Chen, X.P.; Hilton, D.; Rothman, P. Cutting edge: SOCS-1 is a potent inhibitor of IL-4 signal transduction. J. Immunol. 1999, 162, 3770–3774. [Google Scholar] [CrossRef] [PubMed]
  33. Dickensheets, H.L.; Venkataraman, C.; Schindler, U.; Donnelly, R.P. Interferons inhibit activation of STAT6 by interleukin 4 in human monocytes by inducing SOCS-1 gene expression. Proc. Natl. Acad. Sci. USA 1999, 96, 10800–10805. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Pezet, A.; Favre, H.; Kelly, P.A. Inhibition and restoration of prolactin signal transduction by suppressors of cytokine signaling. J. Biol. Chem. 1999, 274, 24497–24502. [Google Scholar] [CrossRef] [Green Version]
  35. Metcalf, D.; Greenhalgh, C.J.; Viney, E.; Willson, T.A.; Starr, R.; Nicola, N.A.; Hilton, D.J.; Alexander, W.S. Gigantism in mice lacking suppressor of cytokine signalling-2. Nature 2000, 405, 1069–1073. [Google Scholar] [CrossRef]
  36. Kopchick, J.J.; Bellush, L.L.; Coschigano, K.T. Transgenic models of growth hormone action. Annu. Rev. Nutr. 1999, 19, 437–446. [Google Scholar] [CrossRef]
  37. Wanke, R.; Milz, S.; Rieger, N.; Ogiolda, L.; Renner-Müller, I.; Brem, G.; Hermanns, W.; Wolf, E. Overgrowth of skin in growth hormone transgenic mice depends on the presence of male gonads. J. Invest. Dermatol. 1999, 113, 967–971. [Google Scholar] [CrossRef] [Green Version]
  38. Sasaki, A.; Yasukawa, H.; Shouda, T.; Kitamura, T.; Dikic, I.; Yoshimura, A. CIS3/SOCS-3 suppresses erythropoietin (EPO) signaling by binding the EPO receptor and JAK2. J. Biol. Chem. 2000, 275, 29338–29347. [Google Scholar] [CrossRef] [Green Version]
  39. Bjørbaek, C.; Elmquist, J.K.; El-Haschimi, K.; Kelly, J.; Ahima, R.S.; Hileman, S.; Flier, J.S. Activation of SOCS-3 messenger ribonucleic acid in the hypothalamus by ciliary neurotrophic factor. Endocrinology 1999, 140, 2035–2043. [Google Scholar] [CrossRef]
  40. Brender, C.; Nielsen, M.; Röpke, C.; Nissen, M.H.; Svejgaard, A.; Billestrup, N.; Geisler, C.; Ødum, N. Interferon-alpha induces transient suppressors of cytokine signalling expression in human T cells. Exp. Clin. Immunogenet. 2001, 18, 80–85. [Google Scholar] [CrossRef]
  41. Dogusan, Z.; Hooghe-Peters, E.L.; Berus, D.; Velkeniers, B.; Hooghe, R. Expression of SOCS genes in normal and leukemic human leukocytes stimulated by prolactin, growth hormone and cytokines. J. Neuroimmunol. 2000, 109, 34–39. [Google Scholar] [CrossRef] [PubMed]
  42. Lee, S.H.; Yun, S.; Piao, Z.H.; Jeong, M.; Kim, D.O.; Jung, H.; Lee, J.; Kim, M.J.; Kim, M.S.; Chung, J.W.; et al. Suppressor of cytokine signaling 2 regulates IL-15-primed human NK cell function via control of phosphorylated Pyk2. J. Immunol. 2010, 185, 917–928. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Sadowski, C.L.; Choi, T.S.; Le, M.; Wheeler, T.T.; Wang, L.H.; Sadowski, H.B. Insulin induction of SOCS-2 and SOCS-3 mRNA expression in C2C12 skeletal muscle cells is mediated by Stat5*. J. Biol. Chem. 2001, 276, 20703–20710. [Google Scholar] [CrossRef] [Green Version]
  44. Harris, J.; Stanford, P.M.; Sutherland, K.; Oakes, S.R.; Naylor, M.J.; Robertson, F.G.; Blazek, K.D.; Kazlauskas, M.; Hilton, H.N.; Wittlin, S.; et al. Socs2 and elf5 mediate prolactin-induced mammary gland development. Mol. Endocrinol. 2006, 20, 1177–1187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Minamoto, S.; Ikegame, K.; Ueno, K.; Narazaki, M.; Naka, T.; Yamamoto, H.; Matsumoto, T.; Saito, H.; Hosoe, S.; Kishimoto, T. Cloning and functional analysis of new members of STAT Induced STAT Inhibitor (SSI) family: SSI-2 and SSI-3. Biochem. Biophys. Res. Comm. 1997, 237, 79–83. [Google Scholar] [CrossRef] [PubMed]
  46. Tannahill, G.M.; Elliott, J.; Barry, A.C.; Hibbert, L.; Cacalano, N.A.; Johnston, J.A. SOCS2 can enhance interleukin-2 (IL-2) and IL-3 signaling by accelerating SOCS3 degradation. Mol. Cell. Biol. 2005, 25, 9115–9126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Nicholson, S.E.; Willson, T.A.; Farley, A.; Starr, R.; Zhang, J.G.; Baca, M.; Alexander, W.S.; Metcalf, D.; Hilton, D.J.; Nicola, N.A. Mutational analyses of the SOCS proteins suggest a dual domain requirement but distinct mechanisms for inhibition of LIF and IL-6 signal transduction. EMBO J. 1999, 18, 375–385. [Google Scholar] [CrossRef] [PubMed]
  48. Goldshmit, Y.; Walters, C.E.; Scott, H.J.; Greenhalgh, C.J.; Turnley, A.M. SOCS2 induces neurite outgrowth by regulation of epidermal growth factor receptor activation. J. Biol. Chem. 2004, 279, 16349–16355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Dey, B.R.; Spence, S.L.; Nissley, P.; Furlanetto, R.W. Interaction of human suppressor of cytokine signaling (SOCS)-2 with the insulin-like growth factor-I receptor. J. Biol. Chem. 1998, 273, 24095–24101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Boisclair, Y.R.; Wang, J.; Shi, J.; Hurst, K.R.; Ooi, G.T. Role of the suppressor of cytokine signaling-3 in mediating the inhibitory effects of interleukin-1beta on the growth hormone-dependent transcription of the acid-labile subunit gene in liver cells. J. Biol. Chem. 2000, 275, 3841–3847. [Google Scholar] [CrossRef]
  51. Cohney, S.J.; Sanden, D.; Cacalano, N.A.; Yoshimura, A.; Mui, A.; Migone, T.S.; Johnston, J.A. SOCS-3 is tyrosine phosphorylated in response to interleukin-2 and suppresses STAT5 phosphorylation and lymphocyte proliferation. Mol. Cell. Biol. 1999, 19, 4980–4988. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Magrangeas, F.; Boisteau, O.; Denis, S.; Jacques, Y.; Minvielle, S. Negative cross-talk between interleukin-3 and interleukin-11 is mediated by suppressor of cytokine signalling-3 (SOCS-3). Biochem. J. 2001, 353, 223–230. [Google Scholar] [CrossRef]
  53. Croker, B.A.; Krebs, D.L.; Zhang, J.G.; Wormald, S.; Willson, T.A.; Stanley, E.G.; Robb, L.; Greenhalgh, C.J.; Förster, I.; Clausen, B.E.; et al. SOCS3 negatively regulates IL-6 signaling in vivo. Nat Immunol 2003, 4, 540–545. [Google Scholar] [CrossRef]
  54. Lejeune, D.; Demoulin, J.B.; Renauld, J.C. Interleukin 9 induces expression of three cytokine signal inhibitors: Cytokine-inducible SH2-containing protein, suppressor of cytokine signalling (SOCS)-2 and SOCS-3, but only SOCS-3 overexpression suppresses interleukin 9 signalling. Biochem. J. 2001, 353, 109–116. [Google Scholar] [CrossRef] [PubMed]
  55. Shen, X.; Hong, F.; Nguyen, V.A.; Gao, B. IL-10 attenuates IFN-alpha-activated STAT1 in the liver: Involvement of SOCS2 and SOCS3. FEBS Lett. 2000, 480, 132–136. [Google Scholar] [CrossRef] [Green Version]
  56. Auernhammer, C.J.; Melmed, S. Interleukin-11 stimulates proopiomelanocortin gene expression and adrenocorticotropin secretion in corticotroph cells: Evidence for a redundant cytokine network in the hypothalamo-pituitary-adrenal axis. Endocrinology 1999, 140, 1559–1566. [Google Scholar] [CrossRef] [PubMed]
  57. Kotenko, S.V.; Izotova, L.S.; Mirochnitchenko, O.V.; Esterova, E.; Dickensheets, H.; Donnelly, R.P.; Pestka, S. Identification, cloning, and characterization of a novel soluble receptor that binds IL-22 and neutralizes its activity. J. Immunol. 2001, 166, 7096–7103. [Google Scholar] [CrossRef] [Green Version]
  58. Adams, T.E.; Hansen, J.A.; Starr, R.; Nicola, N.A.; Hilton, D.J.; Billestrup, N. Growth hormone preferentially induces the rapid, transient expression of SOCS-3, a novel inhibitor of cytokine receptor signaling. J. Biol. Chem. 1998, 273, 1285–1287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Bjørbaek, C.; Elmquist, J.K.; Frantz, J.D.; Shoelson, S.E.; Flier, J.S. Identification of SOCS-3 as a potential mediator of central leptin resistance. Mol. Cell 1998, 1, 619–625. [Google Scholar] [CrossRef] [PubMed]
  60. Faderl, S.; Harris, D.; Van, Q.; Kantarjian, H.M.; Talpaz, M.; Estrov, Z. Granulocyte-macrophage colony-stimulating factor (GM-CSF) induces antiapoptotic and proapoptotic signals in acute myeloid leukemia. Blood 2003, 102, 630–637. [Google Scholar] [CrossRef] [PubMed]
  61. Wang, Q.; Miyakawa, Y.; Fox, N.; Kaushansky, K. Interferon-alpha directly represses megakaryopoiesis by inhibiting thrombopoietin-induced signaling through induction of SOCS-1. Blood 2000, 96, 2093–2099. [Google Scholar] [CrossRef]
  62. Hong, F.; Nguyen, V.A.; Gao, B. Tumor necrosis factor alpha attenuates interferon alpha signaling in the liver: Involvement of SOCS3 and SHP2 and implication in resistance to interferon therapy. FASEB J. 2001, 15, 1595–1597. [Google Scholar] [CrossRef]
  63. Hamanaka, I.; Saito, Y.; Yasukawa, H.; Kishimoto, I.; Kuwahara, K.; Miyamoto, Y.; Harada, M.; Ogawa, E.; Kajiyama, N.; Takahashi, N.; et al. Induction of JAB/SOCS-1/SSI-1 and CIS3/SOCS-3/ SSI-3 is involved in gp130 resistance in cardiovascular system in rat treated with cardiotrophin-1 in vivo. Circ. Res. 2001, 88, 727–732. [Google Scholar] [CrossRef] [Green Version]
  64. Magrangeas, F.; Boisteau, O.; Denis, S.; Jacques, Y.; Minvielle, S. Negative regulation of oncostatin M signaling by suppressor of cytokine signaling (SOCS-3). Eur. Cytokine. Netw. 2001, 12, 309–315. [Google Scholar]
  65. Cacalano, N.A.; Sanden, D.; Johnston, J.A. Tyrosine-phosphorylated SOCS-3 inhibits STAT activation but binds to p120 RasGAP and activates Ras. Nat. Cell Biol. 2001, 3, 460–465. [Google Scholar] [CrossRef]
  66. Park, E.S.; Kim, H.; Suh, J.M.; Park, S.J.; Kwon, O.Y.; Kim, Y.K.; Ro, H.K.; Cho, B.Y.; Chung, J.; Shong, M. Thyrotropin induces SOCS-1 (suppressor of cytokine signaling-1) and SOCS-3 in FRTL-5 thyroid cells. Mol. Endocrinol. 2000, 14, 440–448. [Google Scholar] [CrossRef]
  67. Terstegen, L.; Gatsios, P.; Bode, J.G.; Schaper, F.; Heinrich, P.C.; Graeve, L. The inhibition of interleukin-6-dependent STAT activation by mitogen-activated protein kinases depends on tyrosine 759 in the cytoplasmic tail of glycoprotein 130. J. Biol. Chem. 2000, 275, 18810–18817. [Google Scholar] [CrossRef] [Green Version]
  68. Sobah, M.L.; Liongue, C.; Ward, A.C. SOCS Proteins in Immunity, Inflammatory Diseases, and Immune-Related Cancer. Front. Med. (Lausanne) 2021, 8, 727987. [Google Scholar] [CrossRef]
  69. Chen, Z.; Laurence, A.; Kanno, Y.; Pacher-Zavisin, M.; Zhu, B.M.; Tato, C.; Yoshimura, A.; Hennighausen, L.; O’Shea, J.J. Selective regulatory function of Socs3 in the formation of IL-17-secreting T cells. Proc. Natl. Acad. Sci. USA 2006, 103, 8137–8142. [Google Scholar] [CrossRef] [Green Version]
  70. Brender, C.; Tannahill, G.M.; Jenkins, B.J.; Fletcher, J.; Columbus, R.; Saris, C.J.M.; Ernst, M.; Nicola, N.A.; Hilton, D.J.; Alexander, W.S.; et al. Suppressor of cytokine signaling 3 regulates CD8 T-cell proliferation by inhibition of interleukins 6 and 27. Blood 2007, 110, 2528–2536. [Google Scholar] [CrossRef]
  71. Croker, B.A.; Metcalf, D.; Robb, L.; Wei, W.; Mifsud, S.; DiRago, L.; Cluse, L.A.; Sutherland, K.D.; Hartley, L.; Williams, E.; et al. SOCS3 is a critical physiological negative regulator of G-CSF signaling and emergency granulopoiesis. Immunity 2004, 20, 153–165. [Google Scholar] [CrossRef] [PubMed]
  72. Hansen, J.A.; Lindberg, K.; Hilton, D.J.; Nielsen, J.H.; Billestrup, N. Mechanism of inhibition of growth hormone receptor signaling by suppressor of cytokine signaling proteins. Mol. Endocrinol. 1999, 13, 1832–1843. [Google Scholar] [CrossRef] [PubMed]
  73. Karlsen, A.E.; Rønn, S.G.; Lindberg, K.; Johannesen, J.; Galsgaard, E.D.; Pociot, F.; Nielsen, J.H.; Mandrup-Poulsen, T.; Nerup, J.; Billestrup, N. Suppressor of cytokine signaling 3 (SOCS-3) protects β-cells against interleukin-1β- and interferon-γ-mediated toxicity. Proc. Natl. Acad. Sci. USA 2001, 98, 12191–12196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Zong, C.S.; Chan, J.; Levy, D.E.; Horvath, C.; Sadowski, H.B.; Wang, L.H. Mechanism of STAT3 activation by insulin-like growth factor I receptor. J. Biol. Chem. 2000, 275, 15099–15105. [Google Scholar] [CrossRef] [Green Version]
  75. Emanuelli, B.; Peraldi, P.; Filloux, C.; Sawka-Verhelle, D.; Hilton, D.; Van Obberghen, E. SOCS-3 is an insulin-induced negative regulator of insulin signaling. J. Biol. Chem. 2000, 275, 15985–15991. [Google Scholar] [CrossRef] [Green Version]
  76. Banerjee, A.; Banks, A.S.; Nawijn, M.C.; Chen, X.P.; Rothman, P.B. Cutting edge: Suppressor of cytokine signaling 3 inhibits activation of NFATp. J. Immunol. 2002, 168, 4277–4281. [Google Scholar] [CrossRef] [Green Version]
  77. Aziz, N.; Kim, M.Y.; Cho, J.Y. Anti-inflammatory effects of luteolin: A review of in vitro, in vivo, and in silico studies. J. Ethnopharmacol. 2018, 225, 342–358. [Google Scholar] [CrossRef]
  78. Nicholson, S.E.; Metcalf, D.; Sprigg, N.S.; Columbus, R.; Walker, F.; Silva, A.; Cary, D.; Willson, T.A.; Zhang, J.-G.; Hilton, D.J.; et al. Suppressor of cytokine signaling (SOCS)-5 is a potential negative regulator of epidermal growth factor signaling. Proc. Natl. Acad. Sci. USA 2004, 102, 2328–2333. [Google Scholar] [CrossRef] [Green Version]
  79. Kario, E.; Marmor, M.D.; Adamsky, K.; Citri, A.; Amit, I.; Amariglio, N.; Rechavi, G.; Yarden, Y. Suppressors of cytokine signaling 4 and 5 regulate epidermal growth factor receptor signaling. J. Biol. Chem. 2005, 280, 7038–7048. [Google Scholar] [CrossRef] [Green Version]
  80. Bullock, A.N.; Rodriguez, M.C.; Debreczeni, J.E.; Songyang, Z.; Knapp, S. Structure of the SOCS4-ElonginB/C complex reveals a distinct SOCS box interface and the molecular basis for SOCS-dependent EGFR degradation. Structure 2007, 15, 1493–1504. [Google Scholar] [CrossRef] [Green Version]
  81. Sutherland, J.M.; Keightley, R.A.; Nixon, B.; Roman, S.D.; Robker, R.L.; Russell, D.L.; McLaughlin, E.A. Suppressor of cytokine signaling 4 (SOCS4): Moderator of ovarian primordial follicle activation. J. Cell. Physiol. 2012, 227, 1188–1198. [Google Scholar] [CrossRef]
  82. Lindemann, C.; Hackmann, O.; Delic, S.; Schmidt, N.; Reifenberger, G.; Riemenschneider, M.J. SOCS3 promoter methylation is mutually exclusive to EGFR amplification in gliomas and promotes glioma cell invasion through STAT3 and FAK activation. Acta Neuropathol. 2011, 122, 241–251. [Google Scholar] [CrossRef] [PubMed]
  83. Seki, Y.; Hayashi, K.; Matsumoto, A.; Seki, N.; Tsukada, J.; Ransom, J.; Naka, T.; Kishimoto, T.; Yoshimura, A.; Kubo, M. Expression of the suppressor of cytokine signaling-5 (SOCS5) negatively regulates IL-4-dependent STAT6 activation and Th2 differentiation. Proc. Natl. Acad. Sci. USA 2002, 99, 13003–13008. [Google Scholar] [CrossRef] [Green Version]
  84. Ozaki, A.; Seki, Y.; Fukushima, A.; Kubo, M. The control of allergic conjunctivitis by suppressor of cytokine signaling (SOCS)3 and SOCS5 in a murine model. J. Immunol. 2005, 175, 5489–5497. [Google Scholar] [CrossRef] [Green Version]
  85. Mooney, R.A.; Senn, J.; Cameron, S.; Inamdar, N.; Boivin, L.M.; Shang, Y.; Furlanetto, R.W. Suppressors of cytokine signaling-1 and -6 associate with and inhibit the insulin receptor. A potential mechanism for cytokine-mediated insulin resistance. J. Biol. Chem. 2001, 276, 25889–25893. [Google Scholar] [CrossRef] [Green Version]
  86. Li, L.; Gronning, L.M.; Anderson, P.O.; Li, S.; Edvardsen, K.; Johnston, J.; Kioussis, D.; Shepherd, P.R.; Wang, P. Insulin induces SOCS-6 expression and its binding to the p85 monomer of phosphoinositide 3-kinase, resulting in improvement in glucose metabolism. J. Biol. Chem. 2004, 279, 34107–34114. [Google Scholar] [CrossRef] [Green Version]
  87. Gupta, S.; Mishra, K.; Surolia, A.; Banerjee, K. Suppressor of cytokine signalling-6 promotes neurite outgrowth via JAK2/STAT5-mediated signalling pathway, involving negative feedback inhibition. PLoS ONE 2011, 6, e26674. [Google Scholar] [CrossRef] [Green Version]
  88. Kazi, J.U.; Sun, J.; Phung, B.; Zadjali, F.; Flores-Morales, A.; Rönnstrand, L. Suppressor of cytokine signaling 6 (SOCS6) negatively regulates Flt3 signal transduction through direct binding to phosphorylated tyrosines 591 and 919 of Flt3. J. Biol. Chem. 2012, 287, 36509–36517. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Bayle, J.; Letard, S.; Frank, R.; Dubreuil, P.; De Sepulveda, P. Suppressor of cytokine signaling 6 associates with KIT and regulates KIT receptor signaling. J. Biol. Chem. 2004, 279, 12249–12259. [Google Scholar] [CrossRef] [Green Version]
  90. Choi, Y.B.; Son, M.; Park, M.; Shin, J.; Yun, Y. SOCS-6 negatively regulates T cell activation through targeting p56lck to proteasomal degradation. J. Biol. Chem. 2010, 285, 7271–7280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Matuoka, K.; Miki, H.; Takahashi, K.; Takenawa, T. A novel ligand for an SH3 domain of the adaptor protein Nck bears an SH2 domain and nuclear signaling motifs. Biochem. Biophys. Res. Commun. 1997, 239, 488–492. [Google Scholar] [CrossRef]
  92. Banks, A.S.; Li, J.; McKeag, L.; Hribal, M.L.; Kashiwada, M.; Accili, D.; Rothman, P.B. Deletion of SOCS7 leads to enhanced insulin action and enlarged islets of Langerhans. J. Clin. Invest. 2005, 115, 2462–2471. [Google Scholar] [CrossRef] [Green Version]
  93. Krebs, D.L.; Uren, R.T.; Metcalf, D.; Rakar, S.; Zhang, J.G.; Starr, R.; De Souza, D.P.; Hanzinikolas, K.; Eyles, J.; Connolly, L.M.; et al. SOCS-6 binds to insulin receptor substrate 4, and mice lacking the SOCS-6 gene exhibit mild growth retardation. Mol. Cell. Biol. 2002, 22, 4567–4578. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Tellechea, M.L.; Steinhardt, A.P.; Rodriguez, G.; Taverna, M.J.; Poskus, E.; Frechtel, G. Common variants in SOCS7 gene predict obesity, disturbances in lipid metabolism and insulin resistance. Nutr. Metab. Cardiovasc. Dis. 2013, 23, 424–431. [Google Scholar] [CrossRef]
  95. Polizzoto, M.N.; Bartlett, P.F.; Turnley, A.M. Expression of “suppressor of cytokine signalling” (SOCS) genes in the developing and adult mouse nervous system. J. Comp. Neurol. 2000, 423, 348–358. [Google Scholar] [CrossRef]
  96. Magrangeas, F.; Apiou, F.; Denis, S.; Weidle, U.; Jacques, Y.; Minvielle, S. Cloning and expression of CIS6, chromosome assignment to 3p22 and 2p21 by in situ hybridization. Cytogenet. Cell. Genet. 2000, 88, 78–81. [Google Scholar] [CrossRef]
  97. Liu, X.; Mameza, M.G.; Lee, Y.S.; Eseonu, C.I.; Yu, C.R.; Derwent, J.J.K.; Egwuagu, C.E. Suppressors of cytokine-signaling proteins induce insulin resistance in the retina and promote survival of retinal cells. Diabetes 2008, 57, 1651–1658. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Tanaka, K.; Ichiyama, K.; Hashimoto, M.; Yoshida, H.; Takimoto, T.; Takaesu, G.; Torisu, T.; Hanada, T.; Yasukawa, H.; Fukuyama, S.; et al. Loss of suppressor of cytokine signaling 1 in helper T cells leads to defective Th17 differentiation by enhancing antagonistic effects of IFN-gamma on STAT3 and Smads. J. Immunol. 2008, 180, 3746–3756. [Google Scholar] [CrossRef] [Green Version]
  99. Zhang, Z.; Zeng, B.; Zhang, Z.; Jiao, G.; Li, H.; Jing, Z.; Ouyang, J.; Yuan, X.; Chai, L.; Che, Y.; et al. Suppressor of cytokine signaling 3 promotes bone marrow cells to differentiate into CD8+ T lymphocytes in lung tissue via up-regulating Notch1 expression. Cancer Res. 2009, 69, 1578–1586. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Turnley, A.M.; Starr, R.; Bartlett, P.F. SOCS1 regulates interferon-gamma mediated sensory neuron survival. Neuroreport 2001, 12, 3443–3445. [Google Scholar] [CrossRef] [PubMed]
  101. Cui, M.; Dai, B.; Xin, J.; He, J.; Feng, S. Overexpression of suppressors of cytokine signaling 1 promotes the neuronal differentiation of C17.2. neural stem cells. Cell. Physiol. Biochem. 2014, 33, 528–538. [Google Scholar] [CrossRef] [PubMed]
  102. Cui, M.; Ma, X.L.; Sun, J.; He, J.Q.; Shen, L.; Li, F.G. Overexpression of suppressors of cytokine signaling 1 regulate the proliferation and differentiation of rat-derived neural stem cells. Acta Histochem. 2017, 119, 680–688. [Google Scholar] [CrossRef] [PubMed]
  103. Goldshmit, Y.; Greenhalgh, C.J.; Turnley, A.M. Suppressor of cytokine signalling-2 and epidermal growth factor regulate neurite outgrowth of cortical neurons. Eur. J. Neurosci. 2004, 20, 2260–2266. [Google Scholar] [CrossRef] [PubMed]
  104. Ransome, M.I.; Turnley, A.M. Analysis of neuronal subpopulations in mice over-expressing suppressor of cytokine signaling-2. Neuroscience 2005, 132, 673–687. [Google Scholar] [CrossRef]
  105. Turnley, A.M.; Faux, C.H.; Rietze, R.L.; Coonan, J.R.; Bartlett, P.F. Suppressor of cytokine signalling 2 regulates neuronal differentiation by inhibiting growth hormone signalling. Nat. Neurosci. 2002, 5, 1155–1162. [Google Scholar] [CrossRef]
  106. Scott, H.J.; Stebbing, M.J.; Walters, C.E.; McLenachan, S.; Ransome, M.I.; Nichols, N.R.; Turnley, A.M. Differential effects of SOCS2 on neuronal differentiation and morphology. Brain Res. 2006, 1067, 138–145. [Google Scholar] [CrossRef]
  107. Yadav, A.; Kalita, A.; Dhillon, S.; Banerjee, K. JAK/STAT3 pathway is involved in survival of neurons in response to insulin-like growth factor and negatively regulated by suppressor of cytokine signalling-3. J. Biol. Chem. 2005, 280, 31830–31840. [Google Scholar] [CrossRef] [Green Version]
  108. Mishra, K.K.; Gupta, S.; Banerjee, K. SOCS3 induces neurite differentiation and promotes neuronal cell survival. IUBMB Life 2016, 68, 468–476. [Google Scholar] [CrossRef] [Green Version]
  109. Yoshizumi, T.; Kubo, A.; Murata, H.; Shinonaga, M.; Kanno, H. BC-box motif in SOCS6 induces differentiation of epidermal stem cells into GABAnergic neurons. Int. J. Mol. Sci. 2020, 21, 4947. [Google Scholar] [CrossRef]
  110. Bauer, S. Cytokine control of adult neural stem cells. Ann. N. Y. Acad. Sci. 2009, 1153, 48–56. [Google Scholar] [CrossRef]
  111. Uren, R.T.; Turnley, A.M. Regulation of neurotrophin receptor (Trk) signaling: Suppressor of cytokine signaling 2 (SOCS2) is a new player. Front. Mol. Neurosci. 2014, 7, 39. [Google Scholar] [CrossRef] [PubMed]
  112. Dai, L.; Li, Z.; Liang, W.; Hu, W.; Zhou, S.; Yang, Z.; Tao, Y.; Hou, X.; Xing, Z.; Mao, J.; et al. SOCS proteins and their roles in the development of glioblastoma. Oncol Lett. 2022, 23, 5. [Google Scholar] [CrossRef] [PubMed]
  113. Baker, B.J.; Akhtar, L.N.; Benveniste, E.N. SOCS1 and SOCS3 in the control of CNS immunity. Trends Immunol. 2009, 30, 392–400. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Humphreys, L.M.; Smith, P.; Chen, Z.; Fouad, S.; D’Angiolella, V. The role of E3 ubiquitin ligases in the development and progression of glioblastoma. Cell Death. Differ. 2021, 28, 522–537. [Google Scholar] [CrossRef] [PubMed]
  115. Kaelin, W.G., Jr. Von Hipple-Lindau disease. Annu. Rev. Pathol. Mech. Dis. 2007, 2, 145–173. [Google Scholar] [CrossRef]
  116. Latif, F.; Tory, K.; Gnarra, J.; Yao, M.; Duh, F.M.; Orcutt, M.L.; Stackhouse, T.; Kuzmin, I.; Modi, W.; Geil, L.; et al. Identification of the von Hippel-Lindau disease tumor suppressor gene. Science 1993, 260, 1317–1320. [Google Scholar] [CrossRef]
  117. Pack, S.D.; Zbar, B.; Pak, E.; Ault, D.O.; Humphrey, J.S.; Pham, T.; Hurley, K.; Weil, R.J.; Park, W.S.; Kuzmin, I.; et al. Constitutional von Hippel-Lindau (VHL) gene deletions detected in VHL families by fluorescence in situ hybridization. Cancer Res. 1999, 59, 5560–5564. [Google Scholar] [PubMed]
  118. Iliopoulos, O.; Kibel, A.; Gray, S.; Kaelin, W.G. Tumor suppression by the human von Hippel-Lindau gene product. Nat. Med. 1995, 1, 822–826. [Google Scholar] [CrossRef]
  119. Groulx, I.; Bonicalzi, M.E.; Lee, S. Ran-mediated nuclear export of the von Hippel-Lindau tumor suppressor protein occurs independently of its assembly with cullin-2*. J. Biol. Chem. 2000, 275, 8991–9000. [Google Scholar] [CrossRef] [Green Version]
  120. Groulx, I.; Lee, S. Oxygen-dependent ubiquitination and degradation of hypoxia-inducible factor requires nuclear-cytoplasmic trafficking of the von Hippel-Lindau tumor suppressor protein. Mol. Cell. Biol. 2002, 22, 5319–5336. [Google Scholar] [CrossRef] [Green Version]
  121. Corless, C.L.; Kibel, A.S.; Iliopoulos, O.; Kaelin, W.G. Immunostaining of the von Hippel-Lindau gene product in normal and neoplastic human tissues. Hum. Pathol. 1997, 28, 459–464. [Google Scholar] [CrossRef] [PubMed]
  122. Los, M.; Jansen, G.H.; Kaelin, W.G.; Lips, C.J.; Blijham, G.H.; Voest, E.E. Expression pattern of the von Hippel-Lindau protein in human tissues. Lab. Invest. 1996, 75, 231–238. [Google Scholar] [PubMed]
  123. Ye, Y.; Vasavada, S.; Kuzmin, I.; Stackhouse, T.; Zbar, B.; Williams, B.R. Subcellular localization of the von Hippel-Lindau disease gene product is cell cycle-dependent. Int. J. Cancer 1998, 78, 62–69. [Google Scholar] [CrossRef]
  124. Duan, D.R.; Humphrey, J.S.; Chen, D.Y.; Weng, Y.; Sukegawa, J.; Lee, S.; Gnarra, J.R.; Linehan, W.M.; Klausner, R.D. Characterization of the VHL tumor suppressor gene product: Localization, complex formation, and the effect of natural inactivating mutations. Proc. Natl. Acad. Sci. USA 1995, 92, 6459–6463. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Lee, S.; Neumann, M.; Stearman, R.; Stauber, R.; Pause, A.; Pavlakis, G.N.; Klausner, R.D. Transcription-dependent nuclear-cytoplasmic trafficking is required for the function of the von Hippel-Lindau tumor suppressor protein. Mol. Cell. Biol. 1999, 19, 1486–1497. [Google Scholar] [CrossRef] [Green Version]
  126. Bonicalzi, M.E.; Groulx, I.; de Paulsen, N.; Lee, S. Role of exon 2-encoded β-domain of the von Hippel-Lindau tumor suppressor protein. J. Biol. Chem. 2001, 276, 1407–1416. [Google Scholar] [CrossRef] [Green Version]
  127. Shiao, Y.H.; Resau, J.H.; Nagashima, K.; Anderson, L.M.; Ramakrishna, G. The von Hippel-Lindau tumor suppressor targets to mitochondria. Cancer Res. 2000, 60, 2816–2819. [Google Scholar]
  128. Schoenfeld, A.R.; Davidowitz, E.J.; Burk, R.D. Endoplasmic reticulum/cytosolic localization of von Hippel-Lindau gene products is mediated by a 64-amino acid region. Int. J. Cancer 2001, 91, 457–467. [Google Scholar] [CrossRef]
  129. Stebbins, C.E.; Kaelin, W.G.; Pavletich, N.P. Structure of the VHL-ElonginC-ElonginB complex: Implications for VHL tumor suppressor function. Science 1999, 284, 455–461. [Google Scholar] [CrossRef] [Green Version]
  130. Kibel, A.; Iliopoulos, O.; DeCaprio, J.A.; Kaelin, W.G. Binding of the von Hippel-Lindau tumor suppressor protein to Elongin B and C. Science 1995, 269, 1444–1446. [Google Scholar] [CrossRef]
  131. Lonergan, K.M.; Iliopoulos, O.; Ohh, M.; Kamura, T.; Conaway, R.C.; Kaelin, W.G., Jr. Regulation of hypoxia-inducible mRNAs by the von Hippel-Lindau tumor suppressor protein requires binding to complexes containing elongins B/C and Cul2. Mol. Cell. Biol. 1998, 18, 732–741. [Google Scholar] [CrossRef] [Green Version]
  132. Duan, D.R.; Pause, A.; Burgress, W.H.; Aso, T.; Chen, D.Y.; Garrett, K.P.; Conaway, R.C.; Conaway, J.W.; Linehan, W.M.; Klausner, R.D. Inhibition of transcriptional elongation by the VHL tumor suppressor protein. Science 1995, 269, 1402–1406. [Google Scholar] [CrossRef] [PubMed]
  133. Kamura, T.; Koepp, D.M.; Conrad, M.N.; Skowyra, D.; Moreland, R.J.; Iliopoulos, O.; Lane, W.S.; Kaelin, W.G., Jr. Rbx1, a component of the VHL tumor suppressor complex and SCF ubiquitin ligase. Science 1999, 284, 657–661. [Google Scholar] [CrossRef] [PubMed]
  134. Kamura, T.; Conrad, M.N.; Yan, Q.; Conaway, R.C.; Conaway, J.W. The Rbx1 subunit of SCF and VHL E3 ubiquitin ligase activates Rub1 modification of cullins Cdc53 and Cul2. Genes Dev. 1999, 13, 2928–2933. [Google Scholar] [CrossRef] [PubMed]
  135. Iwai, K.; Yamanaka, K.; Kamura, T.; Minato, N.; Conaway, R.C.; Conaway, J.W.; Klausner, R.D.; Pause, A. Identification of the von Hippel-Lindau tumor-suppressor protein as part of an active E3 ubiquitin ligase complex. Proc. Natl. Acad. Sci. USA 1999, 96, 12436–12441. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Lisztwan, J.; Imbert, G.; Wirbelauer, C.; Gstaiger, M.; Krek, W. The von Hippel-Lindau tumor suppressor protein is a component of an E3 ubiquitin-protein ligase activity. Genes Dev. 1999, 13, 1822–1833. [Google Scholar] [CrossRef] [PubMed]
  137. Maxwell, P.H.; Wiesener, M.S.; Chang, G.W.; Clifford, S.C.; Vaux, E.C.; Cockman, M.E.; Wykoff, C.C.; Pugh, C.W.; Maher, E.R.; Ratcliffe, P.J. The tumour suppressor protein VHL targets hypoxia inducible factors for oxygen-dependent proteolysis. Nature 1999, 399, 271–275. [Google Scholar] [CrossRef] [PubMed]
  138. Ivan, M.; Kondo, K.; Yang, H.; Kim, W.; Valiando, J.; Ohh, M.; Salic, A.; Asara, J.M.; Lane, W.S.; Kaelin, W.G., Jr. HIFalpha targeted for VHL-mediated destruction by proline hydroxylation: Implications for O2 sensing. Science 2001, 292, 464–468. [Google Scholar] [CrossRef]
  139. Jaakkola, P.; Mole, D.R.; Tian, Y.M.; Wilson, M.I.; Gielbert, J.; Gaskell, S.J.; von Kriegsheim, A.; Hebestreit, H.F.; Mukherji, M.; Schofield, C.J.; et al. Targeting of HIF-α to the von Hippel-Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation. Science 2001, 292, 468–472. [Google Scholar] [CrossRef]
  140. Pouysségur, J.; Dayan, F.; Mazure, N.M. Hypoxia signalling in cancer and approaches to enforce tumour regression. Nature 2006, 441, 437–443. [Google Scholar] [CrossRef]
  141. Richards, F.M.; Schofield, P.N.; Fleming, S.; Maher, E.R. Expression of the von Hippel-Lindau disease tumour suppressor gene during human embryogenesis. Hum. Mol. Genet. 1996, 5, 639–644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Nagashima, Y.; Miyagi, Y.; Udagawa, K.; Taki, A.; Misugi, K.; Sakai, N.; Kondo, K.; Kaneko, S.; Yao, M.; Shuin, T. Von Hippel-Lindau tumour suppressor gene. Localization of expression by in situ hybridization. J. Pathol. 1996, 180, 271–274. [Google Scholar] [CrossRef]
  143. Sakashita, N.; Takeya, M.; Kishida, T.; Stackhouse, T.M.; Zbar, B.; Takahashi, K. Expression of von Hippel-Lindau protein in normal and pathological human tissues. Histochem. J. 1999, 31, 133–144. [Google Scholar] [CrossRef]
  144. Kanno, H.; Saljooque, F.; Yamamoto, I.; Hattori, S.; Yao, M.; Shuin, T.; U, H.S. Role of the von Hippel-Lindau tumor suppressor protein during neuronal differentiation. Cancer Res. 2000, 60, 2820–2824. [Google Scholar]
  145. Kanno, H.; Nakano, S.; Kubo, A.; Mimura, T.; Tajima, N.; Sugimoto, N. Neuronal differentiation of neural progenitor cells by intracellular delivery of synthetic oligopeptide derived from Von Hippel-Lindau protein. Protein Pept. Lett. 2009, 16, 1291–1296. [Google Scholar] [CrossRef]
  146. Yamada, H.; Dezawa, M.; Shimazu, S.; Baba, M.; Sawada, H.; Kuroiwa, Y.; Yamamoto, I.; Kanno, H. Transfer of the von Hippel-Lindau gene to neuronal progenitor cells in treatment for Parkinson’s disease. Ann. Neurol. 2003, 54, 352–359. [Google Scholar] [CrossRef]
  147. Josten, F.; Fuss, B.; Feix, M.; Meissner, T.; Hoch, M. Cooperation of JAK/STAT and Notch signaling in the Drosophila foregut. Dev. Biol. 2004, 267, 181–189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Kanno, H.; Xu, Y.; Miyakawa, T.; Kubo, A.; Higashida, T.; Kobayashi, N.B.; Yoshida, T.; Tanokura, M. BC-Box motif-mediated neuronal differentiation of somatic stem cells. Int. J. Mol. Sci. 2018, 19, 466. [Google Scholar] [CrossRef] [Green Version]
  149. Chen, X.; Zhou, B.; Yan, T.; Wu, H.; Feng, J.; Chen, H.; Gao, C.; Peng, T.; Yang, D.; Shen, J. Peroxynitrite enhances self-renewal, proliferation and neuronal differentiation of neural stem/progenitor cells through activating HIF-1α and Wnt/β-catenin signaling pathway. Free Radic. Biol. Med. 2018, 117, 158–167. [Google Scholar] [CrossRef]
  150. Vriend, J.; Reiter, R.J. Melatonin and the von Hippel-Lindau/HIF-1 oxygen sensing mechanism: A review. Biochim. Biophys. Acta (BBA)-Rev. Cancer 2016, 1865, 176–183. [Google Scholar] [CrossRef]
  151. Tanaka, Y.; Kanno, H.; Dezawa, M.; Mimura, T.; Kubo, A.; Yamamoto, I. The role of von Hippel-Lindau protein in the differentiation of neural progenitor cells under normoxic and anoxic conditions. Neurosci. Lett. 2005, 383, 28–32. [Google Scholar] [CrossRef]
  152. Chen, L.; Han, L.; Zhang, K.; Shi, Z.; Zhang, J.; Zhang, A.; Wang, Y.; Song, Y.; Li, Y.; Jiang, T.; et al. VHL regulates the effects of miR-23b on glioma survival and invasion via suppression of HIF-1α/VEGF and β-catenin/Tcf-4 signaling. Neuro Oncol. 2012, 14, 1026–1036. [Google Scholar] [CrossRef] [Green Version]
  153. Sun, X.; Liu, M.; Wei, Y.; Liu, F.; Zhi, X.; Xu, R.; Krissansen, G.W. Overexpression of von Hippel-Lindau tumor suppressor protein and antisense HIF-1alpha eradicates gliomas. Cancer Gene Ther. 2006, 13, 428–435. [Google Scholar] [CrossRef]
  154. Higashida, T.; Jitsuki, S.; Kubo, A.; Mitsushima, D.; Kamiya, Y.; Kanno, H. Skin-derived precursors differentiating into dopaminergic neuronal cells in the brains of Parkinson disease model rats. J. Neurosurg. 2010, 113, 648–655. [Google Scholar] [CrossRef]
  155. Kubo, A.; Yoshida, T.; Kobayashi, N.; Yokoyama, T.; Mimura, T.; Nishiguchi, T.; Higashida, T.; Yamamoto, I.; Kanno, H. Efficient generation of dopamine neuron-like cells from skin-derived precursors with a synthetic peptide derived from von Hippel-Lindau protein. Stem Cells Dev. 2009, 18, 1523–1532. [Google Scholar] [CrossRef]
  156. Maeda, K.; Kanno, H.; Yamazaki, Y.; Kubo, A.; Sato, F.; Yamaguchi, Y.; Saito, T. Transplantation of Von Hippel-Lindau peptide delivered neural stem cells promotes recovery in the injured rat spinal cord. Neuroreport 2009, 20, 1559–1563. [Google Scholar] [CrossRef] [PubMed]
  157. Yamazaki, Y.; Kanno, H.; Maeda, K.; Yoshida, T.; Kobayashi, N.; Kubo, A.; Yamaguchi, Y.; Saito, T. Engrafted VHL peptide-delivered bone marrow stromal cells promote spinal cord repair in rats. Neuroreport 2010, 21, 287–292. [Google Scholar] [CrossRef] [PubMed]
  158. Kanno, H.; Kubo, A.; Yoshizumi, T.; Mikami, T.; Maegawa, J. Isolation of multipotent nestin-expressing stem cells derived from the epidermis of elderly humans and TAT-VHL peptide-mediated neuronal differentiation of these cells. Int. J. Mol. Sci. 2013, 14, 9604–9617. [Google Scholar] [CrossRef]
  159. Ding, X.; Jo, J.; Wang, C.Y.; Cristobal, C.D.; Zuo, Z.; Ye, Q.; Wirianto, M.; Lindeke-Myers, A.; Choi, J.M.; Mohila, C.A.; et al. The Daam2-VHL-Nedd4 axis governs developmental and regenerative oligodendrocyte differentiation. Genes Dev. 2020, 34, 1177–1189. [Google Scholar] [CrossRef]
  160. Cunningham, L.A.; Candelario, K.; Li, L. Roles for HIF-1α in neural stem cell function and the regenerative response to stroke. Behav. Brain Res. 2012, 227, 410–417. [Google Scholar] [CrossRef]
Figure 1. Molecular differentiation pathways of neural stem cells (NSCs). NSCs are maintained by HES1 and 5, ID1 and 3, and the signal transduction and activator of transcription 3 (STAT3). Neuronal differentiation is induced by extracellular factors, such as platelet-derived growth factor (PDGF), brain-derived neurotrophic factor (BDNF), and insulin-like growth factor-1 (IGF-1), and intracellular factors, such as Wnt, Mash, Math, neurogenin1 (Ngn1), von Hippel-Lindau (VHL), and suppressor of cytokine signaling (SOCS). Astrocytic differentiation is induced by extracellular factors, such as interleukin-6 family cytokines (IL-6FCs), leukemia inhibitory factor (LIF), and bone morphogenetic protein (BMP), and intracellular factors, such as Notch, STAT3, and small mother against decapentaplegic (Smad). Oligodendrocytic differentiation is induced by extracellular factors, such as Sonic Hedgehog (Shh), PDGF, fibroblast growth factor (FGF), and bone morphogenetic protein (BMP), and intracellular factors, such as VHL and Olig1 and 2. SOCS and VHL inhibit STAT3 through the Janus kinase–signal transduction and activator of transcription (JAK-STAT) pathway [2,3,4,5,6,7,8,9,10,11,12].
Figure 1. Molecular differentiation pathways of neural stem cells (NSCs). NSCs are maintained by HES1 and 5, ID1 and 3, and the signal transduction and activator of transcription 3 (STAT3). Neuronal differentiation is induced by extracellular factors, such as platelet-derived growth factor (PDGF), brain-derived neurotrophic factor (BDNF), and insulin-like growth factor-1 (IGF-1), and intracellular factors, such as Wnt, Mash, Math, neurogenin1 (Ngn1), von Hippel-Lindau (VHL), and suppressor of cytokine signaling (SOCS). Astrocytic differentiation is induced by extracellular factors, such as interleukin-6 family cytokines (IL-6FCs), leukemia inhibitory factor (LIF), and bone morphogenetic protein (BMP), and intracellular factors, such as Notch, STAT3, and small mother against decapentaplegic (Smad). Oligodendrocytic differentiation is induced by extracellular factors, such as Sonic Hedgehog (Shh), PDGF, fibroblast growth factor (FGF), and bone morphogenetic protein (BMP), and intracellular factors, such as VHL and Olig1 and 2. SOCS and VHL inhibit STAT3 through the Janus kinase–signal transduction and activator of transcription (JAK-STAT) pathway [2,3,4,5,6,7,8,9,10,11,12].
Ijms 24 03880 g001
Figure 2. Structure of suppressor of cytokine signaling (SOCS) and von Hippel-Lindau (VHL) proteins. SOCS family proteins, cytokine-inducible Src homology 2 (SH2)-containing protein (CIS), and SOCS1–7 contain an SH2 domain, BC box, and SOCS box. SOCS1 and SOCS3 also contain a kinase inhibitory region (KIR). VHL contains a β domain, BC box, and VHL box.
Figure 2. Structure of suppressor of cytokine signaling (SOCS) and von Hippel-Lindau (VHL) proteins. SOCS family proteins, cytokine-inducible Src homology 2 (SH2)-containing protein (CIS), and SOCS1–7 contain an SH2 domain, BC box, and SOCS box. SOCS1 and SOCS3 also contain a kinase inhibitory region (KIR). VHL contains a β domain, BC box, and VHL box.
Ijms 24 03880 g002
Figure 3. Molecular mechanism of the suppressor of cytokine signaling (SOCS) proteins. (A) After cytokines bind to its receptor, the Janus kinase–signal transduction and activator of transcription (JAK-STAT) pathway is activated, and the signal transduction and activator of transcription (STAT)-mediated expression of SOCS is induced. In turn, SOCS inhibits the JAK-STAT pathway in a negative feedback loop. (B) SOCS protein recruits Elongin C, Elongin B, Cullin5, Rbx2, and E2 and forms a SOCS/E3 complex. Thereafter, the SOCS/E3 complex binds Janus kinase (JAK); JAK is then ubiquitinated and finally degraded by proteasome 26S.
Figure 3. Molecular mechanism of the suppressor of cytokine signaling (SOCS) proteins. (A) After cytokines bind to its receptor, the Janus kinase–signal transduction and activator of transcription (JAK-STAT) pathway is activated, and the signal transduction and activator of transcription (STAT)-mediated expression of SOCS is induced. In turn, SOCS inhibits the JAK-STAT pathway in a negative feedback loop. (B) SOCS protein recruits Elongin C, Elongin B, Cullin5, Rbx2, and E2 and forms a SOCS/E3 complex. Thereafter, the SOCS/E3 complex binds Janus kinase (JAK); JAK is then ubiquitinated and finally degraded by proteasome 26S.
Ijms 24 03880 g003
Figure 4. Molecular mechanism of von Hippel-Lindau (VHL) protein. (A) Under normoxia, VHL recruits Elongin C, Elongin B, Cullin2, Rbx1, and E2, forming a VHL/E3 complex. Then, VHL binds hypoxia-inducible factor/Janus kinase (HIF/JAK), resulting in HIF/JAK ubiquitination and degradation by proteasome 26S. (B) Under hypoxia, the ubiquitination and degradation of HIF-1α are suppressed; HIF-1α translocates to the nucleus, binds to HIF-1β, binds to the hypoxia response element in the gene promoter, and initiates the transcription of various genes, such as VEGF and EPO [138,139,140].
Figure 4. Molecular mechanism of von Hippel-Lindau (VHL) protein. (A) Under normoxia, VHL recruits Elongin C, Elongin B, Cullin2, Rbx1, and E2, forming a VHL/E3 complex. Then, VHL binds hypoxia-inducible factor/Janus kinase (HIF/JAK), resulting in HIF/JAK ubiquitination and degradation by proteasome 26S. (B) Under hypoxia, the ubiquitination and degradation of HIF-1α are suppressed; HIF-1α translocates to the nucleus, binds to HIF-1β, binds to the hypoxia response element in the gene promoter, and initiates the transcription of various genes, such as VEGF and EPO [138,139,140].
Ijms 24 03880 g004
Figure 5. Double-fluorescence immunocytochemical image of neuronal progenitor cells on day 14 of culture, obtained using a laser scanning confocal microscope. Von Hippel-Lindau (VHL) protein and microtubule-associated proteins (MAPs) are co-expressed in neuronal progenitor cells. VHL is detected with fluorescein isothiocyanate (FITC, green), and MAPs are detected with rhodamine (red). Scale bar = 10 μm.
Figure 5. Double-fluorescence immunocytochemical image of neuronal progenitor cells on day 14 of culture, obtained using a laser scanning confocal microscope. Von Hippel-Lindau (VHL) protein and microtubule-associated proteins (MAPs) are co-expressed in neuronal progenitor cells. VHL is detected with fluorescein isothiocyanate (FITC, green), and MAPs are detected with rhodamine (red). Scale bar = 10 μm.
Ijms 24 03880 g005
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

Kanno, H.; Matsumoto, S.; Yoshizumi, T.; Nakahara, K.; Kubo, A.; Murata, H.; Shuin, T.; U, H.-S. Role of SOCS and VHL Proteins in Neuronal Differentiation and Development. Int. J. Mol. Sci. 2023, 24, 3880. https://doi.org/10.3390/ijms24043880

AMA Style

Kanno H, Matsumoto S, Yoshizumi T, Nakahara K, Kubo A, Murata H, Shuin T, U H-S. Role of SOCS and VHL Proteins in Neuronal Differentiation and Development. International Journal of Molecular Sciences. 2023; 24(4):3880. https://doi.org/10.3390/ijms24043880

Chicago/Turabian Style

Kanno, Hiroshi, Shutaro Matsumoto, Tetsuya Yoshizumi, Kimihiro Nakahara, Atsuhiko Kubo, Hidetoshi Murata, Taro Shuin, and Hoi-Sang U. 2023. "Role of SOCS and VHL Proteins in Neuronal Differentiation and Development" International Journal of Molecular Sciences 24, no. 4: 3880. https://doi.org/10.3390/ijms24043880

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