Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 221
1.
Commun Biol ; 7(1): 629, 2024 May 24.
Article En | MEDLINE | ID: mdl-38789481

Integrin trafficking to and from membrane adhesions is a crucial mechanism that dictates many aspects of a cell's behaviour, including motility, polarisation, and invasion. In endothelial cells (ECs), the intracellular traffic of α5 integrin is regulated by both neuropilin 1 (NRP1) and neuropilin 2 (NRP2), yet the redundancies in function between these co-receptors remain unclear. Moreover, the endocytic complexes that participate in NRP-directed traffic remain poorly annotated. Here we identify an important role for the GTPase-activating protein p120RasGAP in ECs, promoting the recycling of α5 integrin from early endosomes. Mechanistically, p120RasGAP enables transit of endocytosed α5 integrin-NRP1-NRP2 complexes to Rab11+ recycling endosomes, promoting cell polarisation and fibronectin (FN) fibrillogenesis. Silencing of both NRP receptors, or p120RasGAP, resulted in the accumulation of α5 integrin in early endosomes, a loss of α5 integrin from surface adhesions, and attenuated EC polarisation. Endothelial-specific deletion of both NRP1 and NRP2 in the postnatal retina recapitulated our in vitro findings, severely impairing FN fibrillogenesis and polarised sprouting. Our data assign an essential role for p120RasGAP during integrin traffic in ECs and support a hypothesis that NRP receptors co-traffic internalised cargoes. Importantly, we utilise comparative proteomics analyses to isolate a comprehensive map of NRP1-dependent and NRP2-dependent α5 integrin interactions in ECs.


Endosomes , Endothelial Cells , Fibronectins , Integrin alpha5 , Neuropilin-1 , Neuropilin-2 , Proteomics , p120 GTPase Activating Protein , Neuropilin-1/metabolism , Neuropilin-1/genetics , Humans , Integrin alpha5/metabolism , Integrin alpha5/genetics , Endosomes/metabolism , Proteomics/methods , Neuropilin-2/metabolism , Neuropilin-2/genetics , Animals , Fibronectins/metabolism , Endothelial Cells/metabolism , p120 GTPase Activating Protein/metabolism , p120 GTPase Activating Protein/genetics , Protein Transport , Mice , Human Umbilical Vein Endothelial Cells/metabolism , Integrins
2.
Endocr Relat Cancer ; 31(6)2024 Jun 01.
Article En | MEDLINE | ID: mdl-38642579

Neuropilin 2 (NRP2), a transmembrane non-tyrosine kinase receptor, has been described as a potential critical player in the tumourigenesis of several solid cancers and particularly in neuroendocrine neoplasms (NENs). A soluble form of NRP2 (sNRP2) has been previously described and corresponds to a truncated splice isoform. Its prognostic value has never been studied in NEN. NRP2 expression was studied by immunochemistry on tissue microarrays (n = 437) and on circulating tumour cells (CTCs, n = 5 patients with neuroendocrine carcinoma, NEC). We described the levels of sNRP2 in 229 patients with NEN using the ELISA method to identify the factors associated with sNRP2 levels and to evaluate its prognostic role; 90 blood donors represented the healthy control group. NRP2 was found in 97% of neuroendocrine tumours (396/410) and in 74% of NEC (20/27). NRP2 was also expressed in CTC of all the studied patients. The receiver operating characteristic (ROC) analysis showed that sNRP2 had a weak capacity to discriminate between NEN patients and healthy controls (area under curve (AUC) = 0.601, P = 0.053). Abnormal sNRP2 levels were associated with inflammatory syndrome, bone and peritoneal metastases, and abnormal chromogranin A levels. Patients with high sNRP2 levels (sNRP2Q3-Q4) had significantly poorer overall survival in multivariate analysis (HR 0.16, 95% CI (0.04-0.67), P = 0.015). In conclusion, the present study found that sNRP2 and NRP2 could represent a new prognostic biomarker and a therapeutic target, respectively, particularly in aggressive NEN.


Biomarkers, Tumor , Neuroendocrine Tumors , Neuropilin-2 , Humans , Female , Neuropilin-2/metabolism , Neuropilin-2/genetics , Male , Middle Aged , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/pathology , Neuroendocrine Tumors/blood , Aged , Adult , Biomarkers, Tumor/metabolism , Prognosis , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Aged, 80 and over , Young Adult
3.
J Pathol ; 262(2): 175-188, 2024 02.
Article En | MEDLINE | ID: mdl-37946610

Neuropilin-2 (NRP2) is a multifunctional protein engaged in the regulation of angiogenesis, lymphangiogenesis, axon guidance, and tumor metastasis, but its function in colitis remains unclear. Here, we found that NRP2 was an inflammation-sensing protein rapidly and dramatically induced in myeloid cells, especially in macrophages, under inflammatory contexts. NRP2 deficiency in myeloid cells exacerbated dextran sulfate sodium salt-induced experimental colitis by promoting polarization of M1 macrophages and colon injury. Mechanistically, NRP2 could be induced via NF-κB activation by TNF-α in macrophages, but exerted an inhibitory effect on NF-κB signaling, forming a negative feedback loop with NF-κB to sense and alleviate inflammation. Deletion of NRP2 in macrophages broke this negative feedback circuit, leading to NF-κB overactivation, inflammatory exacerbation, and more severe colitis. Collectively, these findings reveal inflammation restriction as a role for NRP2 in macrophages under inflammation contexts and suggest that NRP2 in macrophages may relieve inflammation in inflammatory bowel disease. © 2023 The Pathological Society of Great Britain and Ireland.


Colitis , NF-kappa B , Humans , Animals , Mice , NF-kappa B/metabolism , Neuropilin-2/genetics , Neuropilin-2/metabolism , Colitis/pathology , Inflammation/pathology , Macrophages/pathology , Dextran Sulfate/toxicity , Dextran Sulfate/metabolism , Mice, Inbred C57BL , Disease Models, Animal
4.
Horm Metab Res ; 55(10): 701-710, 2023 Oct.
Article En | MEDLINE | ID: mdl-37813099

Circular RNAs (circRNAs) are implicated in regulating the pathogenesis of papillary thyroid carcinoma (PTC). Herein, we aimed to investigate how circRNA phosphatidylinositol 4-kinase IIIα (circPI4KA, hsa_circ_0062389) functioned as an oncogene in PTC. CircPI4KA, microRNA-1287-5p (miR-1287-5p) and Neuropilin-2 (NRP2) level detection were completed by reverse transcription-quantitative polymerase chain reaction assay. Cell proliferation was assessed through Cell Counting Kit-8 assay, colony formation assay, and EdU assay. Transwell assay was used for detecting migration and invasion abilities. Cell migration was also determined by wound healing assay. Cell apoptosis was assessed using flow cytometry assay. The protein examination was performed using western blot. Glycolysis was evaluated via commercial kits. Dual-luciferase reporter assay and RNA immunoprecipitation assay were conducted for target analysis. The role of circPI4KA in vivo was explored and analyzed via tumor xenograft assay. CircPI4KA was significantly upregulated in PTC tissues and cells. Knockdown of circPI4KA suppressed proliferation, migration, invasion, glycolysis, and induced apoptosis of PTC cells. CircPI4KA interacted with miR-1287-5p in PTC cells. The antitumor function of circPI4KA downregulation was reversed by inhibition of miR-1287-5p. The miR-1287-5p directly targeted NRP2, and circPI4KA elevated the NRP2 expression by sponging miR-1287-5p. PTC progression was impeded by miR-1287-5p via targeting NRP2. Silencing circPI4KA inhibited tumor growth in vivo through the miR-1287-5p/NRP2 axis. The collective results revealed that circPI4KA induced the upregulation of NRP2 via sponging miR-1287-5p, thus acting as a carcinogenic factor in PTC.


MicroRNAs , Neuropilin-2 , RNA, Circular , Thyroid Neoplasms , Humans , Carcinogenesis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Glycolysis/genetics , MicroRNAs/genetics , Neuropilin-2/genetics , Thyroid Cancer, Papillary/genetics , Thyroid Neoplasms/genetics , RNA, Circular/genetics
5.
Front Endocrinol (Lausanne) ; 14: 1223021, 2023.
Article En | MEDLINE | ID: mdl-37600714

Introduction: Neuropilin 2 (NRP2) mediates the effects of class 3 semaphorins and vascular endothelial growth factor and is implicated in axonal guidance and angiogenesis. Moreover, NRP2 expression is suggested to be involved in the regulation of bone homeostasis. Indeed, osteoblasts and osteoclasts express NRP2 and male and female global Nrp2 knockout mice have a reduced bone mass accompanied by reduced osteoblast and increased osteoclast counts. Methods: We first examined the in vitro effect of the calciotropic hormone 1,25-dihydroxyvitamin D3 [1,25(OH)2D3] on Nrp2 transcription in osteoblasts. We next generated mice with a conditional deletion of Nrp2 in the osteoblast cell lineage under control of the paired related homeobox 1 promoter and mice with a conditional Nrp2 knockdown in osteoclasts under control of the Lysozyme promoter. Mice were examined under basal conditions or after treatment with either the bone anabolic vitamin D3 analog WY 1048 or with 1,25(OH)2D3. Results and discussion: We show that Nrp2 expression is induced by 1,25(OH)2D3 in osteoblasts and is associated with enrichment of the vitamin D receptor in an intronic region of the Nrp2 gene. In male mice, conditional deletion of Nrp2 in osteoblast precursors and mature osteoblasts recapitulated the bone phenotype of global Nrp2 knockout mice, with a reduced cortical cross-sectional tissue area and lower trabecular bone content. However, female mice with reduced osteoblastic Nrp2 expression display a reduced cross-sectional tissue area but have a normal trabecular bone mass. Treatment with the vitamin D3 analog WY 1048 (0.4 µg/kg/d, 14 days, ip) resulted in a similar increase in bone mass in both genotypes and genders. Deleting Nrp2 from the osteoclast lineage did not result in a bone phenotype, even though in vitro osteoclastogenesis of hematopoietic cells derived from mutant mice was significantly increased. Moreover, treatment with a high dose of 1,25(OH)2D3 (0.5 µg/kg/d, 6 days, ip), to induce osteoclast-mediated bone resorption, resulted in a similar reduction in trabecular and cortical bone mass. In conclusion, osteoblastic Nrp2 expression is suggested to regulate bone homeostasis in a sex-specific manner.


Cancellous Bone , Neuropilin-2 , Osteoblasts , Animals , Female , Male , Mice , Cholecalciferol , Cross-Sectional Studies , Neuropilin-2/genetics , Vascular Endothelial Growth Factor A , Calcitriol
6.
Sci Transl Med ; 15(694): eade5855, 2023 05 03.
Article En | MEDLINE | ID: mdl-37134151

Prostate cancers are largely unresponsive to immune checkpoint inhibitors (ICIs), and there is strong evidence that programmed death-ligand 1 (PD-L1) expression itself must be inhibited to activate antitumor immunity. Here, we report that neuropilin-2 (NRP2), which functions as a vascular endothelial growth factor (VEGF) receptor on tumor cells, is an attractive target to activate antitumor immunity in prostate cancer because VEGF-NRP2 signaling sustains PD-L1 expression. NRP2 depletion increased T cell activation in vitro. In a syngeneic model of prostate cancer that is resistant to ICI, inhibition of the binding of VEGF to NRP2 using a mouse-specific anti-NRP2 monoclonal antibody (mAb) resulted in necrosis and tumor regression compared with both an anti-PD-L1 mAb and control immunoglobulin G. This therapy also decreased tumor PD-L1 expression and increased immune cell infiltration. We observed that the NRP2, VEGFA, and VEGFC genes are amplified in metastatic castration-resistant and neuroendocrine prostate cancer. We also found that individuals with NRP2High PD-L1High metastatic tumors had lower androgen receptor expression and higher neuroendocrine prostate cancer scores than other individuals with prostate cancer. In organoids derived from patients with neuroendocrine prostate cancer, therapeutic inhibition of VEGF binding to NRP2 using a high-affinity humanized mAb suitable for clinical use also diminished PD-L1 expression and caused a substantial increase in immune-mediated tumor cell killing, consistent with the animal studies. These findings provide justification for the initiation of clinical trials using this function-blocking NRP2 mAb in prostate cancer, especially for patients with aggressive disease.


Prostatic Neoplasms , Vascular Endothelial Growth Factor A , Male , Animals , Humans , Vascular Endothelial Growth Factor A/metabolism , Neuropilin-2/genetics , Neuropilin-2/metabolism , Signal Transduction , B7-H1 Antigen/genetics , Prostatic Neoplasms/metabolism
7.
Elife ; 122023 04 03.
Article En | MEDLINE | ID: mdl-37010951

Secreted semaphorin 3F (Sema3F) and semaphorin 3A (Sema3A) exhibit remarkably distinct effects on deep layer excitatory cortical pyramidal neurons; Sema3F mediates dendritic spine pruning, whereas Sema3A promotes the elaboration of basal dendrites. Sema3F and Sema3A signal through distinct holoreceptors that include neuropilin-2 (Nrp2)/plexinA3 (PlexA3) and neuropilin-1 (Nrp1)/PlexA4, respectively. We find that Nrp2 and Nrp1 are S-palmitoylated in cortical neurons and that palmitoylation of select Nrp2 cysteines is required for its proper subcellular localization, cell surface clustering, and also for Sema3F/Nrp2-dependent dendritic spine pruning in cortical neurons, both in vitro and in vivo. Moreover, we show that the palmitoyl acyltransferase ZDHHC15 is required for Nrp2 palmitoylation and Sema3F/Nrp2-dependent dendritic spine pruning, but it is dispensable for Nrp1 palmitoylation and Sema3A/Nrp1-dependent basal dendritic elaboration. Therefore, palmitoyl acyltransferase-substrate specificity is essential for establishing compartmentalized neuronal structure and functional responses to extrinsic guidance cues.


Semaphorins , Semaphorins/metabolism , Semaphorin-3A/metabolism , Neuropilin-2/genetics , Neuropilin-2/metabolism , Lipoylation , Neurons/metabolism , Neuropilin-1/genetics , Neuropilin-1/metabolism
8.
J Cosmet Dermatol ; 22(8): 2333-2342, 2023 Aug.
Article En | MEDLINE | ID: mdl-37025072

BACKGROUND: Exosomes (Exos) and their contained microRNAs (miRNAs) have been emergingly recognized as key regulators in spanning biological processes, including proliferation and angiogenesis. AIM OF THE STUDY: This work investigates the function of Exos derived from adipose-derived mesenchymal stem cells (adMSCs) in viability of keloid fibroblasts (KFs). METHODS: Abnormally expressed miRNAs in keloid tissues were screened using the GEO dataset GSE113620. Meanwhile, miRNAs enriched in adMSC-Exos were predicted by bioinformatics system. Exos were extracted from acquired adMSCs and identified, which were co-incubated with KFs. Uptake of Exos by KFs was examined by fluorescence staining. Viability, proliferation, and apoptosis of KFs were analyzed by CCK-8, EdU labeling, and TUNEL assays. Conditioned medium of KFs was collected to stimulate angiogenesis of human umbilical vein endothelial cells (HUVECs). Binding between miR-7846-3p and neuropilin 2 (NRP2) was validated by luciferase assay. Protein levels of NRP2 and the Hedgehog pathway molecules were analyzed by western blot analysis. RESULTS: miR-7846-3p was predicted as an exosomal miRNA aberrantly expressed in keloids. AdMSC-Exos reduced viability, proliferation, and apoptosis resistance of KFs, and they blocked the angiogenesis of HUVECs. miR-7846-3p targeted NRP2 mRNA. miR-7846-3p upregulation in KFs suppressed NRP2 expression and reduced the expression of Hedgehog pathway molecules SHH, SMO, and GLI1. Either miR-7846-3p inhibition in Exos or NRP2 overexpression in KFs blocked the effects of Exos and restored the viability, proliferation, and pro-angiogenic role of KFs. CONCLUSION: This work unravels that adMSC-Exos-derived miR-7846-3p suppresses NRP2 and inactivates the Hedgehog signaling to reduce proliferation and pro-angiogenic role of KFs.


Keloid , Mesenchymal Stem Cells , MicroRNAs , Humans , Cell Proliferation/genetics , Endothelial Cells , Fibroblasts/metabolism , Hedgehog Proteins/metabolism , Keloid/genetics , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , Neuropilin-2/genetics , Neuropilin-2/metabolism
9.
Curr Mol Pharmacol ; 16(7): 787-799, 2023.
Article En | MEDLINE | ID: mdl-36545728

BACKGROUND: MicroRNAs (miRs) are small noncoding RNAs that are crucial in the development and progression of tumours. Melanoma is an aggressive form of skin cancer and is resistant to most of the chemotherapeutic agents. However, the role of miRs in melanoma remains poorly studied. OBJECTIVE: The work aimed to demonstrate that miR-331-3p is downregulated in melanoma against the benign melanocytic nevi. METHODS: RT-PCR analysis was performed for the expression of proteins; cell proliferation and wound healing assays were carried out. Flow cytometry study was conducted for cell cycle analysis; colony formation assay was performed by soft agar method. For developing a tumour xenograft model, nu/nu mice were selected. RESULTS: Up-regulation of miR-331-3p in melanoma cells decreased cell proliferation, cell migration, and also drug resistance. Over-expression of miR-331-3p resulted in suppression of NRP2 and up-regulation of E-cadherin levels. Moreover, the levels of MDR1, ABCG-2, and ABCG-5 were decreased. However, the knockdown of NRP2 demonstrated similar effects as that of miR- 331-3p overexpression in tumour cells. Overexpression of miR-331-3p caused significant inhibition of tumour growth and its metastasis in mice model of melanoma, which was associated with depletion of NRP2 protein and increased expression of E-cadherin. However, the effects of miR- 331-3p on the migration, cell proliferation, and self-renewal were overturned by the upregulation of NRP2, which also resulted in the inhibition of E-cadherin and overexpression of MDR-1, ABCG-2, and ABCG-5. CONCLUSION: The findings point out the key role of miR-331-3p in the progression and drug resistance of melanoma involving NRP2.


Melanoma , MicroRNAs , Animals , Mice , Humans , Neuropilin-2/genetics , Neuropilin-2/metabolism , Cell Line, Tumor , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , Cell Movement/genetics , Drug Resistance , Cadherins/metabolism , Cell Proliferation , Gene Expression Regulation, Neoplastic
10.
Med Oncol ; 40(1): 62, 2022 Dec 26.
Article En | MEDLINE | ID: mdl-36567417

Lung cancer is the leading cause of malignant tumor-related deaths worldwide. The presence of tumor-initiating cells in lung cancer leads to tumor recurrence, metastasis, and resistance to conventional treatment. Cleavage and polyadenylation specificity factor 4 (CPSF4) activation in tumor cells contributes to the poor prognosis of lung cancer. However, the precise biological functions and molecular mechanisms of CPSF4 in the regulation of tumor-initiating cells remain unclear. We demonstrated that CPSF4 promotes tumor-initiating phenotype and confers chemoresistance to paclitaxel both in vitro and in vivo. Mechanistically, we showed that CPSF4 binds to the promoters of vascular endothelial growth factor (VEGF) and neuropilin-2 (NRP2) and activated their transcription. In addition, we showed that CPSF4/VEGF/NRP2-mediated tumor-initiating phenotype and chemoresistance through TAZ induction. Furthermore, analysis of clinical data revealed that lung cancer patients with high CPSF4 expression exhibit high expression levels of VEGF, NRP2, and TAZ and that expression of these proteins are positively correlated with poor prognosis. Importantly, selective inhibition of VEGF, NRP2, or TAZ markedly suppressed CPSF4-mediated tumor-initiating phenotype and chemoresistance. Our findings reveal the mechanism of CPSF4 modulating tumor-initiating phenotype and chemoresistance in lung cancer and indicate that the CPSF4-VEGF-NRP2-TAZ signaling pathway may be a prognosis marker and therapeutic target in lung cancer.


Carcinogenesis , Cleavage And Polyadenylation Specificity Factor , Gene Expression Regulation, Neoplastic , Lung Neoplasms , Neuropilin-2 , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Vascular Endothelial Growth Factor A , Humans , Cell Line, Tumor , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Neuropilin-2/genetics , Phenotype , Signal Transduction , Vascular Endothelial Growth Factor A/genetics , Cleavage And Polyadenylation Specificity Factor/metabolism , Carcinogenesis/genetics
11.
Pharmacol Res ; 185: 106517, 2022 Nov.
Article En | MEDLINE | ID: mdl-36265554

Regulatory B cells (Bregs) potently suppress immune disorders, including allergic contact hypersensitivity (CHS). IKKß overactivation is prominent in various inflammatory diseases. However, its effect on Bregs has not been defined. This study is to investigate the new regulator and inhibitory mechanism of Bregs. IkkßC46A transgenic mice with a Cys46 mutation, resulting in increased IKKß activation, were employed for analysis. IL-10-competent CD9+ Bregs were expanded in IkkßC46A mice and B cell specific-IkkßC46A mutation mice. IkkßC46A mutant CD9+ Bregs had stronger suppressive effects on CD4+ and CD8+ T cells in vitro and CHS responses in vivo. The inhibitory CD9+ Bregs from IkkßC46A mice were characterized by upregulated Neuropilin 2 (Nrp2) and IL-10 in comparison with that of Ikkßwt mice. Interestingly, increased expression of Nrp2 was observed in CD9+ Bregs compared with that of CD9- B cells in wild-type mice. The suppressive activity of wild-type CD9+ Bregs in vitro was attenuated by inhibition of Nrp2 on Bregs or silencing its ligand Sema3f on CD4+ T cells. Our findings delineate a distinct role of IKKß activation in enhancing Bregs to disturb the immune balance. It identifies Nrp2 as a novel regulatory molecule of Bregs that partly contributes to B cell-mediated immune tolerance.


B-Lymphocytes, Regulatory , Immune System Diseases , Animals , Mice , CD8-Positive T-Lymphocytes/metabolism , I-kappa B Kinase/metabolism , Immune System Diseases/metabolism , Interleukin-10 , Mice, Inbred C57BL , Mice, Transgenic , Neuropilin-2/genetics , Neuropilin-2/metabolism
12.
Reprod Biol ; 22(4): 100697, 2022 Dec.
Article En | MEDLINE | ID: mdl-36306654

Endometriosis is a gynecological disorder seriously affecting the health and life of women of reproductive age. Neuropilin 2 (NRP2) has been indicated to display a high level in ectopic endometrium. Nevertheless, the specific function of NRP2 in endometriosis is unanswered. RT-qPCR was utilized to detect expression of NRP2 and SMAD family member 2 (SMAD2) in endometrial tissues or endometrial stromal cells (ESCs). Protein levels of transforming growth factor beta (TGF-ß) signaling-associated markers and epithelial-mesenchymal transition (EMT)-related markers were examined by western blotting. Transwell assays were utilized for detecting the impact of NRP2 on ectopic ESC phenotypes. ChIP and luciferase reporter assays were performed for identification of the relationship between NRP2 and SMAD2. In this study, NRP2 was overexpressed in ectopic endometria in comparison to eutopic endometria. Depletion of NRP2 restrained ectopic ESC migration, invasiveness and EMT. TGF-ß signaling-mediated activation of SMAD2 transcriptionally upregulated NRP2 expression in ectopic ESCs. TGF-ß treatment could rescue NRP2 silencing-induced suppressive impact on the behaviors of ectopic ESCs. Overall, the activation of TGF-ß signaling contributes to the migration and invasiveness of ectopic ESCs by targeting NRP2.


Endometriosis , Neuropilin-2 , Transforming Growth Factor beta , Female , Humans , Cell Movement/genetics , Endometriosis/metabolism , Endometrium/metabolism , Neuropilin-2/genetics , Neuropilin-2/metabolism , Stromal Cells/metabolism , Transforming Growth Factor beta/metabolism
13.
Int J Mol Sci ; 23(15)2022 Jul 29.
Article En | MEDLINE | ID: mdl-35955539

Neuropilin 1 (NRP1) represents one of the two homologous neuropilins (NRP, splice variants of neuropilin 2 are the other) found in all vertebrates. It forms a transmembrane glycoprotein distributed in many human body tissues as a (co)receptor for a variety of different ligands. In addition to its physiological role, it is also associated with various pathological conditions. Recently, NRP1 has been discovered as a coreceptor for the SARS-CoV-2 viral entry, along with ACE2, and has thus become one of the COVID-19 research foci. However, in addition to COVID-19, the current review also summarises its other pathological roles and its involvement in clinical diseases like cancer and neuropathic pain. We also discuss the diversity of native NRP ligands and perform a joint analysis. Last but not least, we review the therapeutic roles of NRP1 and introduce a series of NRP1 modulators, which are typical peptidomimetics or other small molecule antagonists, to provide the medicinal chemistry community with a state-of-the-art overview of neuropilin modulator design and NRP1 druggability assessment.


COVID-19 , Neoplasms , Animals , Humans , Neuropilin-1/chemistry , Neuropilin-1/genetics , Neuropilin-2/genetics , SARS-CoV-2
14.
Oncogene ; 41(37): 4307-4317, 2022 09.
Article En | MEDLINE | ID: mdl-35986103

Neuroendocrine prostate cancer (NEPC), a lethal subset of prostate cancer, is characterized by loss of AR signaling and resulting resistance to AR-targeted therapy during neuroendocrine transdifferentiation, for which the molecular mechanisms remain unclear. Here, we report that neuropilin 2 (NRP2) is upregulated in both de novo and therapy-induced NEPC, which induces neuroendocrine markers, neuroendocrine cell morphology, and NEPC cell aggressive behavior. NRP2 silencing restricted NEPC tumor xenograft growth. Mechanistically, NRP2 engages in reciprocal crosstalk with AR, where NRP2 is transcriptionally inhibited by AR, and in turn suppresses AR signaling by downregulating the AR transcriptional program and confers resistance to enzalutamide. Moreover, NRP2 physically interacts with VEGFR2 through the intracellular SEA domain to activate STAT3 phosphorylation and subsequently SOX2, thus driving NEPC differentiation and growth. Collectively, these results characterize NRP2 as a driver of NEPC and suggest NRP2 as a potential therapeutic target in NEPC.


Carcinoma, Neuroendocrine , Prostatic Neoplasms , Carcinoma, Neuroendocrine/genetics , Cell Line, Tumor , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Male , Neuropilin-2/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology
15.
Am J Pathol ; 192(11): 1592-1603, 2022 11.
Article En | MEDLINE | ID: mdl-35985479

Appropriate coordination of smooth muscle contraction and relaxation is essential for normal colonic motility. The impact of perturbed motility ranges from moderate, in conditions such as colitis, to potentially fatal in the case of pseudo-obstruction. The mechanisms underlying aberrant motility and the extent to which they can be targeted pharmacologically are incompletely understood. This study identified colonic smooth muscle as a major site of expression of neuropilin 2 (Nrp2) in mice and humans. Mice with inducible smooth muscle-specific knockout of Nrp2 had an increase in evoked contraction of colonic rings in response to carbachol at 1 and 4 weeks following initiation of deletion. KCl-induced contractions were also increased at 4 weeks. Colonic motility was similarly enhanced, as evidenced by faster bead expulsion in Nrp2-deleted mice versus Nrp2-intact controls. In length-tension analysis of the distal colon, passive tension was similar in Nrp2-deficient and Nrp2-intact mice, but at low strains, active stiffness was greater in Nrp2-deficient animals. Consistent with the findings in conditional Nrp2 mice, Nrp2-null mice showed increased contractility in response to carbachol and KCl. Evaluation of selected proteins implicated in smooth muscle contraction revealed no significant differences in the level of α-smooth muscle actin, myosin light chain, calponin, or RhoA. Together, these findings identify Nrp2 as a novel regulator of colonic contractility that may be targetable in conditions characterized by dysmotility.


Colon , Gastrointestinal Motility , Muscle Contraction , Muscle, Smooth , Neuropilin-2 , Animals , Humans , Mice , Carbachol/pharmacology , Colon/metabolism , Colon/physiology , Mice, Knockout , Muscle Contraction/drug effects , Muscle Contraction/genetics , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Neuropilin-2/genetics , Neuropilin-2/metabolism , Gastrointestinal Motility/drug effects , Gastrointestinal Motility/genetics
16.
Front Immunol ; 13: 830169, 2022.
Article En | MEDLINE | ID: mdl-35651620

Tumor-associated macrophages (TAMs) exert profound influence over breast cancer progression, promoting immunosuppression, angiogenesis, and metastasis. Neuropilin-2 (NRP2), consisting of the NRP2a and NRP2b isoforms, is a co-receptor for heparin-binding growth factors including VEGF-C and Class 3 Semaphorins. Selective upregulation in response to environmental stimuli and independent signaling pathways endow the NRP2 isoforms with unique functionality, with NRP2b promoting increased Akt signaling via receptor tyrosine kinases including VEGFRs, MET, and PDGFR. Although NRP2 has been shown to regulate macrophage/TAM biology, the role of the individual NRP2a/NRP2b isoforms in TAMs has yet to be evaluated. Using transcriptional profiling and spectral flow cytometry, we show that NRP2 isoform expression was significantly higher in TAMs from murine mammary tumors. NRP2a/NRP2b levels in human breast cancer metastasis were dependent upon the anatomic location of the tumor and significantly correlated with TAM infiltration in both primary and metastatic breast cancers. We define distinct phenotypes of NRP2 isoform-expressing TAMs in mouse models of breast cancer and within malignant pleural effusions from breast cancer patients which were exclusive of neuropilin-1 expression. Genetic depletion of either NRP2 isoform in macrophages resulted in a dramatic reduction of LPS-induced IL-10 production, defects in phagosomal processing of apoptotic breast cancer cells, and increase in cancer cell migration following co-culture. By contrast, depletion of NRP2b, but not NRP2a, inhibited production of IL-6. These results suggest that NRP2 isoforms regulate both shared and unique functionality in macrophages and are associated with distinct TAM subsets in breast cancer.


Breast Neoplasms , Neuropilin-2 , Animals , Breast Neoplasms/pathology , Female , Humans , Mice , Neuropilin-1/genetics , Neuropilin-2/genetics , Neuropilin-2/metabolism , Protein Isoforms , Tumor-Associated Macrophages
17.
Oncogene ; 41(30): 3747-3760, 2022 07.
Article En | MEDLINE | ID: mdl-35754042

Aberrant transcriptional activity of androgen receptor (AR) is one of the dominant mechanisms for developing of castration-resistant prostate cancer (CRPC). Analyzing AR-transcriptional complex related to CRPC is therefore important towards understanding the mechanism of therapy resistance. While studying its mechanism, we observed that a transmembrane protein called neuropilin-2 (NRP2) plays a contributory role in forming a novel AR-transcriptional complex containing nuclear pore proteins. Using immunogold electron microscopy, high-resolution confocal microscopy, chromatin immunoprecipitation, proteomics, and other biochemical techniques, we delineated the molecular mechanism of how a specific splice variant of NRP2 becomes sumoylated upon ligand stimulation and translocates to the inner nuclear membrane. This splice variant of NRP2 then stabilizes the complex between AR and nuclear pore proteins to promote CRPC specific gene expression. Both full-length and splice variants of AR have been identified in this specific transcriptional complex. In vitro cell line-based assays indicated that depletion of NRP2 not only destabilizes the AR-nuclear pore protein interaction but also inhibits the transcriptional activities of AR. Using an in vivo bone metastasis model, we showed that the inhibition of NRP2 led to the sensitization of CRPC cells toward established anti-AR therapies such as enzalutamide. Overall, our finding emphasize the importance of combinatorial inhibition of NRP2 and AR as an effective therapeutic strategy against treatment refractory prostate cancer.


Prostatic Neoplasms, Castration-Resistant , Androgens/pharmacology , Cell Line, Tumor , Humans , Male , Neuropilin-2/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Signal Transduction
18.
Mol Cancer Res ; 20(8): 1208-1221, 2022 08 05.
Article En | MEDLINE | ID: mdl-35533267

Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal types of cancer, as it commonly metastasizes to the liver resulting in an overall poor prognosis. However, the molecular mechanism involved in liver metastasis remains poorly understood. Here, we aimed to identify the MUC16-mediated molecular mechanism of PDAC-liver metastasis. Previous studies demonstrated that MUC16 and its C-terminal (Cter) domain are involved in the aggressiveness of PDAC. In this study, we observed MUC16 and its Cter expression significantly high in human PDAC tissues, PDAC organoids, and metastatic liver tissues, while no expression was observed in normal pancreatic tissues using IHC and immunofluorescence (IFC) analyses. MUC16 knockdown in SW1990 and CD18/HPAF PDAC cells significantly decreased the colony formation, migration, and endothelial/p-selectin binding. In contrast, MUC16-Cter ectopic overexpression showed significantly increased colony formation and motility in MiaPaCa2 pancreatic cancer cells. Interestingly, MUC16 promoted cell survival and colonization in the liver, mimicking an ex vivo environment. Furthermore, MUC16 enhanced liver metastasis in the in vivo mouse model. Our integrated analyses of RNA-sequencing suggested that MUC16 alters Neuropilin-2 (NRP2) and cell adhesion molecules in pancreatic cancer cells. Furthermore, we identified that MUC16 regulated NRP2 via JAK2/STAT1 signaling in PDAC. NRP2 knockdown in MUC16-overexpressed PDAC cells showed significantly decreased cell adhesion and migration. Overall, the findings indicate that MUC16 regulates NRP2 and induces metastasis in PDAC. IMPLICATIONS: This study shows that MUC16 plays a critical role in PDAC liver metastasis by mediating NRP2 regulation by JAK2/STAT1 axis, thereby paving the way for future therapy efforts for metastatic PDAC.


Adenocarcinoma , Carcinoma, Pancreatic Ductal , Liver Neoplasms , Neuropilin-2 , Pancreatic Neoplasms , Adenocarcinoma/pathology , Animals , CA-125 Antigen/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Adhesion , Cell Line, Tumor , Cell Movement , Cell Proliferation , Humans , Liver Neoplasms/genetics , Liver Neoplasms/secondary , Membrane Proteins/metabolism , Mice , Neoplasm Metastasis , Neuropilin-2/genetics , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms
19.
Bioengineered ; 13(4): 11042-11049, 2022 04.
Article En | MEDLINE | ID: mdl-35473507

To investigate the effects of silencing neuropilin-2(NRP-2) on the proliferation, migration, and invasion of colorectal cancer(CRC) HT-29. Lipofectamine 2000 was used to transfect specific siRNA for NRP-2 and nonspecific control siRNA into human colorectal cancer HT-29 as the transfection group and meaningless sequence group. HT-29 cultured in a medium was used as the blank control group. The expression levels of NRP-2 mRNA in the cells were detected by real-time fluorescence quantitative PCR. The expressions of proliferation-associated protein Ki-67 in the cells were detected by immunochemical staining. Migration ability was assessed by a monolayer cell scratch wound damage and repair experiment. The Transwell chamber invasion experiment was adopted to determine invasive ability by measuring the number of tumor cells crossing the chamber membrane. Compared with the meaningless sequence group and blank control group, real-time fluorescence quantitative PCR showed that the relative expression level of NRP-2 mRNA in the transfection group was significantly decreased(P < 0.05). Results of immunochemical staining revealed that the expression of Ki-67 protein in the transfected cells was significantly reduced, and the proliferation ability was decreased(P < 0.05). The results further showed that the scratch healing rate of the transfected cells decreased after 24 h of healing(P < 0.05). Results of Transwell invasion assay showed that the number of cells passing through the stromal membrane of the upper chamber to the back of the chamber was significantly reduced in the transfection group(p < 0.05). Silencing NRP-2 could inhibit the proliferation, migration, and invasion of colorectal cancer HT-29.


Colorectal Neoplasms , Neuropilin-2 , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , HT29 Cells , Humans , Ki-67 Antigen/metabolism , Neoplasm Invasiveness , Neuropilin-2/genetics , Neuropilin-2/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
20.
Epigenomics ; 14(3): 153-162, 2022 02.
Article En | MEDLINE | ID: mdl-35021853

Smoking could predispose individuals to a more severe COVID-19 by upregulating a particular gene known as mdig, which is mediated through a number of well-known histone modifications. Smoking might regulate the transcription-activating H3K4me3 mark, along with the transcription-repressing H3K9me3 and H3K27me3 marks, in a way to favor SARS-CoV-2 entry by enhancing the expression of ACE2, NRP1 and NRP2, AT1R, CTSD and CTSL, PGE2 receptors 2-4, SLC6A20 and IL-6, all of which interact either directly or indirectly with important receptors, facilitating viral entry in COVID-19.


Lay abstract The role of smoking in development of several respiratory diseases has been clearly established. A significant proportion of these deleterious effects is mediated through epigenetic mechanisms, particularly histone modifications. Recent evidence indicates that smoking induces the expression of a mediator known as mdig, which in turn alters the transcription of several key proteins that have been implicated in development of COVID-19.


COVID-19/genetics , Dioxygenases/genetics , Epigenesis, Genetic , Histone Demethylases/genetics , Histones/genetics , Nuclear Proteins/genetics , Protein Processing, Post-Translational , Smoking/genetics , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/metabolism , COVID-19/diagnosis , COVID-19/metabolism , COVID-19/virology , Cathepsin D/genetics , Cathepsin D/metabolism , Cathepsin L/genetics , Cathepsin L/metabolism , Dioxygenases/metabolism , Histone Demethylases/metabolism , Histones/metabolism , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Methylation , Neuropilin-1/genetics , Neuropilin-1/metabolism , Neuropilin-2/genetics , Neuropilin-2/metabolism , Nuclear Proteins/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E/metabolism , Risk Factors , SARS-CoV-2/genetics , SARS-CoV-2/growth & development , SARS-CoV-2/metabolism , Smoking/metabolism , Smoking/pathology , Virus Internalization
...