Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.602
Filter
1.
Int J Mol Sci ; 25(15)2024 Jul 28.
Article in English | MEDLINE | ID: mdl-39125815

ABSTRACT

Neurological symptoms associated with COVID-19, acute and long term, suggest SARS-CoV-2 affects both the peripheral and central nervous systems (PNS/CNS). Although studies have shown olfactory and hematogenous invasion into the CNS, coinciding with neuroinflammation, little attention has been paid to susceptibility of the PNS to infection or to its contribution to CNS invasion. Here we show that sensory and autonomic neurons in the PNS are susceptible to productive infection with SARS-CoV-2 and outline physiological and molecular mechanisms mediating neuroinvasion. Our infection of K18-hACE2 mice, wild-type mice, and golden Syrian hamsters, as well as primary peripheral sensory and autonomic neuronal cultures, show viral RNA, proteins, and infectious virus in PNS neurons, satellite glial cells, and functionally connected CNS tissues. Additionally, we demonstrate, in vitro, that neuropilin-1 facilitates SARS-CoV-2 neuronal entry. SARS-CoV-2 rapidly invades the PNS prior to viremia, establishes a productive infection in peripheral neurons, and results in sensory symptoms often reported by COVID-19 patients.


Subject(s)
COVID-19 , Neuropilin-1 , SARS-CoV-2 , Animals , SARS-CoV-2/physiology , SARS-CoV-2/pathogenicity , COVID-19/virology , COVID-19/pathology , COVID-19/metabolism , Mice , Neuropilin-1/metabolism , Neuropilin-1/genetics , Viremia/virology , Central Nervous System/virology , Central Nervous System/pathology , Central Nervous System/metabolism , Sensory Receptor Cells/virology , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/pathology , Mesocricetus , Humans , Angiotensin-Converting Enzyme 2/metabolism , Mice, Inbred C57BL , Virus Internalization , Male
2.
Int J Mol Sci ; 25(15)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39126095

ABSTRACT

Olfactory perception is an important physiological function for human well-being and health. Loss of olfaction, or anosmia, caused by viral infections such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has received considerable attention, especially in persistent cases that take a long time to recover. This review discusses the integration of different components of the olfactory epithelium to serve as a structural and functional unit and explores how they are affected during viral infections, leading to the development of olfactory dysfunction. The review mainly focused on the role of receptors mediating the disruption of olfactory signal transduction pathways such as angiotensin converting enzyme 2 (ACE2), transmembrane protease serine type 2 (TMPRSS2), neuropilin 1 (NRP1), basigin (CD147), olfactory, transient receptor potential vanilloid 1 (TRPV1), purinergic, and interferon gamma receptors. Furthermore, the compromised function of the epithelial sodium channel (ENaC) induced by SARS-CoV-2 infection and its contribution to olfactory dysfunction are also discussed. Collectively, this review provides fundamental information about the many types of receptors that may modulate olfaction and participate in olfactory dysfunction. It will help to understand the underlying pathophysiology of virus-induced anosmia, which may help in finding and designing effective therapies targeting molecules involved in viral invasion and olfaction. To the best of our knowledge, this is the only review that covered all the receptors potentially involved in, or mediating, the disruption of olfactory signal transduction pathways during COVID-19 infection. This wide and complex spectrum of receptors that mediates the pathophysiology of olfactory dysfunction reflects the many ways in which anosmia can be therapeutically managed.


Subject(s)
Anosmia , COVID-19 , SARS-CoV-2 , Humans , COVID-19/metabolism , COVID-19/complications , COVID-19/physiopathology , COVID-19/virology , Anosmia/physiopathology , Anosmia/etiology , Anosmia/metabolism , Angiotensin-Converting Enzyme 2/metabolism , Olfactory Mucosa/metabolism , Olfactory Mucosa/virology , Signal Transduction , Serine Endopeptidases/metabolism , Neuropilin-1/metabolism , Basigin/metabolism , TRPV Cation Channels/metabolism
3.
Pediatr Surg Int ; 40(1): 221, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39133317

ABSTRACT

PURPOSE: The aim of this study was to detect candidate oncogenes of rhabdoid tumor of the kidney (RTK) and evaluate their roles in RTK in vitro. METHODS: An integrated analysis of messenger RNA (mRNA) and microRNA (miRNA) sequencing was performed to determine the expression profile of exosome-derived miRNAs and mRNAs in human RTK-derived cell lines and a human embryonic renal cell line. A Gene Ontology enrichment analysis was performed to analyze the functional characteristics of differentially expressed mRNAs in RTK cells. Matrigel invasion and wound-healing assays were performed to evaluate the cell invasion and migration abilities. RESULTS: Forty mRNAs were highly expressed in RTK cells targeted by exosomal miRNAs, the expression of which was lower in RTK cells than in the controls. These mRNAs were primarily related to cell adhesion. Of these mRNAs, we selected neuropilin 1 (NRP1) as a candidate oncogene because its upregulated expression is associated with a poor prognosis of several types of tumors. RTK cells in which NRP1 had been knocked down exhibited decreased invasive and migratory abilities. CONCLUSION: Our study indicates that NRP1 acts as an oncogene by promoting the invasion and migration of RTK cells and that it could serve as a therapeutic target.


Subject(s)
Cell Movement , Kidney Neoplasms , Neoplasm Invasiveness , Neuropilin-1 , Rhabdoid Tumor , Humans , Neuropilin-1/genetics , Neuropilin-1/metabolism , Cell Movement/genetics , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Neoplasm Invasiveness/genetics , Rhabdoid Tumor/genetics , Rhabdoid Tumor/pathology , Cell Line, Tumor , RNA, Messenger/genetics , RNA, Messenger/metabolism , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Gene Knockdown Techniques/methods
4.
Breast Cancer Res ; 26(1): 122, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138514

ABSTRACT

BACKGROUND: A better understanding of ductal carcinoma in situ (DCIS) is urgently needed to identify these preinvasive lesions as distinct clinical entities. Semaphorin 3F (SEMA3F) is a soluble axonal guidance molecule, and its coreceptors Neuropilin 1 (NRP1) and NRP2 are strongly expressed in invasive epithelial BC cells. METHODS: We utilized two cell line models to represent the progression from a healthy state to the mild-aggressive or ductal carcinoma in situ (DCIS) stage and, ultimately, to invasive cell lines. Additionally, we employed in vivo models and conducted analyses on patient databases to ensure the translational relevance of our results. RESULTS: We revealed SEMA3F as a promoter of invasion during the DCIS-to-invasive ductal carcinoma transition in breast cancer (BC) through the action of NRP1 and NRP2. In epithelial cells, SEMA3F activates epithelialmesenchymal transition, whereas it promotes extracellular matrix degradation and basal membrane and myoepithelial cell layer breakdown. CONCLUSIONS: Together with our patient database data, these proof-of-concept results reveal new SEMA3F-mediated mechanisms occurring in the most common preinvasive BC lesion, DCIS, and represent potent and direct activation of its transition to invasion. Moreover, and of clinical and therapeutic relevance, the effects of SEMA3F can be blocked directly through its coreceptors, thus preventing invasion and keeping DCIS lesions in the preinvasive state.


Subject(s)
Breast Neoplasms , Carcinoma, Intraductal, Noninfiltrating , Neoplasm Invasiveness , Nerve Tissue Proteins , Neuropilin-1 , Neuropilin-2 , Humans , Neuropilin-1/metabolism , Neuropilin-1/genetics , Female , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Neuropilin-2/metabolism , Neuropilin-2/genetics , Carcinoma, Intraductal, Noninfiltrating/metabolism , Carcinoma, Intraductal, Noninfiltrating/pathology , Carcinoma, Intraductal, Noninfiltrating/genetics , Cell Line, Tumor , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Epithelial-Mesenchymal Transition/genetics , Animals , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mice , Carcinoma, Ductal, Breast/pathology , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/genetics , Gene Expression Regulation, Neoplastic , Signal Transduction
5.
J Gastric Cancer ; 24(3): 300-315, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38960889

ABSTRACT

PURPOSE: Gastric cancer (GC) is among the deadliest malignancies and the third leading cause of cancer-related deaths worldwide. Galectin-1 (Gal-1) is a primary protein secreted by cancer-associated fibroblasts (CAFs); however, its role and mechanisms of action of Gal-1 in GC remain unclear. In this study, we stimulated GC cells with exogenous human recombinant galectin-1 protein (rhGal-1) to investigate its effects on the proliferation, migration, and resistance to cisplatin. MATERIALS AND METHODS: We used simulated rhGal-1 protein as a paracrine factor produced by CAFs to induce GC cells and investigated its promotional effects and mechanisms in GC progression and cisplatin resistance. Immunohistochemical (IHC) assay confirmed that Gal-1 expression was associated with clinicopathological parameters and correlated with the expression of neuropilin-1 (NRP-1), c-JUN, and Wee1. RESULTS: Our study reveals Gal-1 expression was significantly associated with poor outcomes. Gal-1 boosts the proliferation and metastasis of GC cells by activating the NRP-1/C-JUN/Wee1 pathway. Gal-1 notably increases GC cell resistance to cisplatin The NRP-1 inhibitor, EG00229, effectively counteracts these effects. CONCLUSIONS: These findings revealed a potential mechanism by which Gal-1 promotes GC growth and contributes to chemoresistance, offering new therapeutic targets for the treatment of GC.


Subject(s)
Cell Proliferation , Cisplatin , Drug Resistance, Neoplasm , Galectin 1 , Neuropilin-1 , Stomach Neoplasms , Stomach Neoplasms/pathology , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Stomach Neoplasms/genetics , Humans , Galectin 1/genetics , Galectin 1/metabolism , Cisplatin/pharmacology , Cisplatin/therapeutic use , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Neuropilin-1/metabolism , Neuropilin-1/genetics , Cell Proliferation/drug effects , Male , Female , Disease Progression , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Signal Transduction/drug effects , Middle Aged , Mice , Animals , Cell Movement/drug effects , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/pathology
6.
BMC Urol ; 24(1): 147, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39014364

ABSTRACT

BACKGROUND: Insulin-like growth factor 2 mRNA-binding protein 2 (IGF2BP2) has been confirmed to play oncogenic role in many cancers. However, the role and mechanism of IGF2BP2 in bladder cancer (BCa) still deserves to be further revealed. METHODS: The mRNA and protein levels of IGF2BP2 and neuronilin-1 (NRP1) were detected by real-time quantitative PCR (RT-qPCR) and western blot. Cell proliferation, apoptosis, migration and invasion were determined using colony formation assay, EdU assay, CCK8 assay, flow cytometry and transwell assay. Xenograft tumor model was conducted to evaluate the role of IGF2BP2 in vivo. THP-1-M0 macrophages were co-cultured with the condition medium (CM) of BCa cells to induce polarization. M2 macrophage polarization was assessed by detecting the mRNA levels of M2 macrophage markers using RT-qPCR and measuring the proportion of M2 macrophage markers using flow cytometry. Moreover, MeRIP and RIP assay were performed to assess m6A level and the interaction between IGF2BP2 and NRP1. RESULTS: IGF2BP2 and NRP1 were upregulated in BCa tissues and cells. IGF2BP2 knockdown suppressed BCa cell growth and metastasis, as well as inhibited BCa tumor growth. After THP-1-M0 macrophages were co-cultured with the CM of BCa cells, the levels of M2 macrophage markers were markedly enhanced, while this effect was abolished by IGF2BP2 knockdown. IGF2BP2 level was positively correlated with NRP1 level, and it could increase NRP1 mRNA stability. NRP1 overexpression reversed the suppressive effect of IGF2BP2 knockdown on M2 macrophage polarization and BCa cell progression. CONCLUSION: m6A-reader IGF2BP2 enhanced M2 macrophage polarization and BCa cell progression by promoting NRP1 mRNA stability.


Subject(s)
Macrophages , Neuropilin-1 , RNA, Messenger , RNA-Binding Proteins , Urinary Bladder Neoplasms , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/metabolism , Humans , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Macrophages/metabolism , RNA, Messenger/metabolism , Neuropilin-1/genetics , Neuropilin-1/metabolism , Mice , Gene Expression Regulation, Neoplastic , Animals , Cell Polarity/physiology , Cell Line, Tumor
7.
Signal Transduct Target Ther ; 9(1): 169, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38956074

ABSTRACT

More than 90% of hepatocellular carcinoma (HCC) cases develop in the presence of fibrosis or cirrhosis, making the tumor microenvironment (TME) of HCC distinctive due to the intricate interplay between cancer-associated fibroblasts (CAFs) and cancer stem cells (CSCs), which collectively regulate HCC progression. However, the mechanisms through which CSCs orchestrate the dynamics of the tumor stroma during HCC development remain elusive. Our study unveils a significant upregulation of Sema3C in fibrotic liver, HCC tissues, peripheral blood of HCC patients, as well as sorafenib-resistant tissues and cells, with its overexpression correlating with the acquisition of stemness properties in HCC. We further identify NRP1 and ITGB1 as pivotal functional receptors of Sema3C, activating downstream AKT/Gli1/c-Myc signaling pathways to bolster HCC self-renewal and tumor initiation. Additionally, HCC cells-derived Sema3C facilitated extracellular matrix (ECM) contraction and collagen deposition in vivo, while also promoting the proliferation and activation of hepatic stellate cells (HSCs). Mechanistically, Sema3C interacted with NRP1 and ITGB1 in HSCs, activating downstream NF-kB signaling, thereby stimulating the release of IL-6 and upregulating HMGCR expression, consequently enhancing cholesterol synthesis in HSCs. Furthermore, CAF-secreted TGF-ß1 activates AP1 signaling to augment Sema3C expression in HCC cells, establishing a positive feedback loop that accelerates HCC progression. Notably, blockade of Sema3C effectively inhibits tumor growth and sensitizes HCC cells to sorafenib in vivo. In sum, our findings spotlight Sema3C as a novel biomarker facilitating the crosstalk between CSCs and stroma during hepatocarcinogenesis, thereby offering a promising avenue for enhancing treatment efficacy and overcoming drug resistance in HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Semaphorins , Tumor Microenvironment , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Humans , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Tumor Microenvironment/genetics , Semaphorins/genetics , Semaphorins/metabolism , Integrin beta1/genetics , Integrin beta1/metabolism , Mice , Signal Transduction/genetics , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Neuropilin-1/genetics , Neuropilin-1/metabolism , Cell Line, Tumor , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/metabolism , Animals , Gene Expression Regulation, Neoplastic/genetics , Sorafenib/pharmacology , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Disease Progression
8.
Nat Commun ; 15(1): 5731, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38977708

ABSTRACT

Neuropilin-1 (NRP1), a co-receptor for various cytokines, including TGF-ß, has been identified as a potential therapeutic target for fibrosis. However, its role and mechanism in renal fibrosis remains elusive. Here, we show that NRP1 is upregulated in distal tubular (DT) cells of patients with transplant renal insufficiency and mice with renal ischemia-reperfusion (I-R) injury. Knockout of Nrp1 reduces multiple endpoints of renal injury and fibrosis. We find that Nrp1 facilitates the binding of TNF-α to its receptor in DT cells after renal injury. This signaling results in a downregulation of lysine crotonylation of the metabolic enzyme Cox4i1, decreases cellular energetics and exacerbation of renal injury. Furthermore, by single-cell RNA-sequencing we find that Nrp1-positive DT cells secrete collagen and communicate with myofibroblasts, exacerbating acute kidney injury (AKI)-induced renal fibrosis by activating Smad3. Dual genetic deletion of Nrp1 and Tgfbr1 in DT cells better improves renal injury and fibrosis than either single knockout. Together, these results reveal that targeting of NRP1 represents a promising strategy for the treatment of AKI and subsequent chronic kidney disease.


Subject(s)
Acute Kidney Injury , Fibrosis , Mice, Knockout , Neuropilin-1 , Receptor, Transforming Growth Factor-beta Type I , Reperfusion Injury , Smad3 Protein , Neuropilin-1/metabolism , Neuropilin-1/genetics , Animals , Humans , Mice , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Acute Kidney Injury/genetics , Receptor, Transforming Growth Factor-beta Type I/metabolism , Receptor, Transforming Growth Factor-beta Type I/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Smad3 Protein/metabolism , Smad3 Protein/genetics , Male , Tumor Necrosis Factor-alpha/metabolism , Signal Transduction , Mice, Inbred C57BL , Kidney Tubules/pathology , Kidney Tubules/metabolism , Myofibroblasts/metabolism , Myofibroblasts/pathology , Collagen/metabolism
9.
Sci Rep ; 14(1): 17513, 2024 07 30.
Article in English | MEDLINE | ID: mdl-39080306

ABSTRACT

Cancer treatment with vascular disrupting agents (VDAs) causes rapid and extensive necrosis in solid tumors. However, these agents fall short in eliminating all malignant cells, ultimately leading to tumor regrowth. Here, we investigated whether the molecular changes in the tumor microenvironment induced by VDA treatment sensitize the tumors for secondary nanotherapy enhanced by clinical-stage tumor penetrating peptide iRGD. Treatment of peritoneal carcinomatosis (PC) and breast cancer mice with VDA combretastatin A-4 phosphate (CA4P) resulted in upregulation of the iRGD receptors αv-integrins and NRP-1, particularly in the peripheral tumor tissue. In PC mice treated with CA4P, coadministration of iRGD resulted in an approximately threefold increase in tumor accumulation and a more homogenous distribution of intraperitoneally administered nanoparticles. Notably, treatment with a combination of CA4P, iRGD, and polymersomes loaded with a novel anthracycline Utorubicin (UTO-PS) resulted in a significant decrease in the overall tumor burden in PC-bearing mice, while avoiding overt toxicities. Our results indicate that VDA-treated tumors can be targeted therapeutically using iRGD-potentiated nanotherapy and warrant further studies on the sequential targeting of VDA-induced molecular signatures.


Subject(s)
Nanoparticles , Tumor Microenvironment , Animals , Tumor Microenvironment/drug effects , Mice , Female , Nanoparticles/chemistry , Bibenzyls/pharmacology , Bibenzyls/chemistry , Cell Line, Tumor , Humans , Stilbenes/pharmacology , Stilbenes/administration & dosage , Oligopeptides/chemistry , Oligopeptides/pharmacology , Neuropilin-1/metabolism , Peritoneal Neoplasms/drug therapy , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage
10.
J Phys Chem B ; 128(29): 7141-7147, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39010661

ABSTRACT

The binding of the virus to host cells is the first step in viral infection. Human cell angiotensin converting enzyme 2 (ACE2) is the most popular receptor for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), while other receptors have recently been observed in experiments. Neuropilin-1 protein (NRP1) is one of them, but the mechanism of its binding to the wild type (WT) and different variants of the virus remain unclear at the atomic level. In this work, all-atom umbrella sampling simulations were performed to clarify the binding mechanism of NRP1 to the spike protein fragments 679-685 of the WT, Delta, and Omicron BA.1 variants. We found that the Delta variant binds most strongly to NRP1, while the affinity for Omicron BA.1 slightly decreases for NRP1 compared to that of WT, and the van der Waals interaction plays a key role in stabilizing the studied complexes. The change in the protonation state of the His amino acid results in different binding free energies between variants. Consistent with the experiment, decreasing the pH was shown to increase the binding affinity of the virus to NRP1. Our results indicate that Delta and Omicron mutations not only affect fusogenicity but also affect NRP1 binding. In addition, we argue that viral evolution does not further improve NRP1 binding affinity which remains in the µM range but may increase immune evasion.


Subject(s)
Molecular Dynamics Simulation , Neuropilin-1 , Protein Binding , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Neuropilin-1/metabolism , Neuropilin-1/chemistry , Humans , SARS-CoV-2/chemistry , SARS-CoV-2/metabolism , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/metabolism , COVID-19/virology , COVID-19/metabolism , Angiotensin-Converting Enzyme 2/metabolism , Angiotensin-Converting Enzyme 2/chemistry
11.
Cell Host Microbe ; 32(6): 945-946, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38870904

ABSTRACT

In this issue of Cell Host & Microbe, Shang et al. identify murine neuropilin 1 as a host factor that binds reovirus particles, directing cell entry and contributing to viral dissemination and neurovirulence. This study highlights the reovirus model system to investigate host receptors and their significance in viral pathogenesis.


Subject(s)
Neurons , Neuropilin-1 , Reoviridae , Virus Internalization , Animals , Mice , Neurons/virology , Neuropilin-1/metabolism , Reoviridae/physiology , Reoviridae/genetics , Reoviridae/pathogenicity , Humans , Host-Pathogen Interactions , Reoviridae Infections/virology , Receptors, Virus/metabolism
12.
Pharmacol Res ; 205: 107259, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38871237

ABSTRACT

The osteopontin-derived peptide FOL-005 stimulates hair growth. Using ligand-receptor glyco-capture technology we identified neuropilin-1 (NRP-1), a known co-receptor for vascular endothelial growth factor (VEGF) receptors, as the most probable receptor for FOL-005 and the more stable analogue FOL-026. X-ray diffraction and microscale thermophoresis analysis revealed that FOL-026 shares binding site with VEGF in the NRP-1 b1-subdomain. Stimulation of human umbilical vein endothelial cells with FOL-026 resulted in phosphorylation of VEGFR-2, ERK1/2 and AKT, increased cell growth and migration, stimulation of endothelial tube formation and inhibition of apoptosis in vitro. FOL-026 also promoted angiogenesis in vivo as assessed by subcutaneous Matrigel plug and hind limb ischemia models. NRP-1 knock-down or treatment of NRP-1 antagonist EG00229 blocked the stimulatory effects of FOL-026 on endothelial cells. Exposure of human coronary artery smooth muscle cells to FOL-026 stimulated cell growth, migration, inhibited apoptosis, and induced VEGF gene expression and VEGFR-2/AKT phosphorylation by an NRP-1-dependent mechanism. RNA sequencing showed that FOL-026 activated pathways involved in tissue repair. These findings identify NRP-1 as the receptor for FOL-026 and show that its biological effects mimic that of growth factors binding to the VEGF receptor family. They also suggest that FOL-026 may have therapeutical potential in conditions that require vascular repair and/or enhanced angiogenesis.


Subject(s)
Human Umbilical Vein Endothelial Cells , Neovascularization, Physiologic , Neuropilin-1 , Osteopontin , Neuropilin-1/metabolism , Humans , Human Umbilical Vein Endothelial Cells/drug effects , Animals , Neovascularization, Physiologic/drug effects , Osteopontin/metabolism , Osteopontin/genetics , Cell Movement/drug effects , Vascular Endothelial Growth Factor Receptor-2/metabolism , Cell Proliferation/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Male , Peptides/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Apoptosis/drug effects , Mice, Inbred C57BL , Protein Binding , Ischemia/drug therapy , Ischemia/metabolism , Mice , Angiogenesis
13.
Biochem Pharmacol ; 226: 116336, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38844264

ABSTRACT

The pathological mechanisms underlying the sex-dependent presentation of calcific aortic stenosis (AS) remain poorly understood. We aim to analyse sex-specific responses of valve interstitial cells (VICs) to calcific environments and to identify new pathological and potentially druggable targets. First, VICs from stenotic patients were modelled using pro-calcifying media (HP). Both male and female VICs were inflamed upon calcific HP challenge, although the inflammatory response was higher in female VICs. The osteogenic and calcification responses were higher in male VICs. To identify new players involved in the responses to HP, proteomics analyses were performed on additional calcifying VICs. Neuropilin-1 (NRP-1) was significantly up-regulated in male calcifying VICs and that was confirmed in aortic valves (AVs), especially nearby neovessels and calcifications. Regardless of the sex, NRP-1 expression was correlated to inflammation, angiogenesis and osteogenic markers, but with stronger associations in male AVs. To further evidence the role of NRP-1, in vitro experiments of silencing or supplementation with soluble NRP-1 (sNRP-1) were performed. NRP-1 silencing or addition of sNRP-1 reduced/mended the expression of any sex-specific response triggered by HP. Moreover, NRP-1 regulation contributed to significantly diminish the baseline enhanced expression of pro-inflammatory, pro-angiogenic and pro-osteogenic markers mainly in male VICs. Validation studies were conducted in stenotic AVs. In summary, pharmacologic targeting of NRP-1 could be used to target sex-specific phenotypes in AS as well as to exert protective effects by reducing the basal expression of pathogenic markers only in male VICs.


Subject(s)
Aortic Valve Stenosis , Aortic Valve , Calcinosis , Neuropilin-1 , Osteogenesis , Male , Female , Neuropilin-1/metabolism , Neuropilin-1/genetics , Humans , Osteogenesis/drug effects , Osteogenesis/physiology , Calcinosis/metabolism , Calcinosis/pathology , Calcinosis/genetics , Aortic Valve Stenosis/metabolism , Aortic Valve Stenosis/pathology , Aortic Valve/pathology , Aortic Valve/metabolism , Sex Characteristics , Inflammation/metabolism , Inflammation/pathology , Aged , Cells, Cultured , Phenotype , Middle Aged , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology
14.
Life Sci ; 351: 122764, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38838817

ABSTRACT

The discovery of SARS-CoV-2 RNA in the periodontal tissues of patients who tested positive for COVID-19, 24 days post the initial symptom onset, indicates the oral cavity could serve as a viral reservoir. This research aims to investigate the antiviral capabilities of Ovatodiolide, introducing a novel periodontal ligament organoid model for the study of SARS-CoV-2. We have successfully established a reliable and expandable organoid culture from the human periodontal ligament, showcasing characteristics typical of epithelial stem cells. This organoid model enables us to delve into the lesser-known aspects of dental epithelial stem cell biology and their interactions with viruses and oral tissues. We conducted a series of in vitro and ex vivo studies to examine the inhibitory impacts of Ova on SARS-CoV-2. Our findings indicate that Ovatodiolide molecules can bind effectively to the NRP1 active domain. Our study identifies potential interaction sites for Ovatodiolide (OVA) within the b1 domain of the NRP1 receptor. We generated point mutations at this site, resulting in three variants: Y25A, T44A, and a double mutation Y25A/T44A. While these mutations did not alter the binding activity of the spike protein, they did impact the concentration of OVA required for inhibition. The inhibitory concentrations for these variants are 15 µM for Y25A, 15.2 µM for T44A, and 25 µM for the double mutant Y25A/T44A. In addition, in vitro inhibition experiments demonstrate that the EC50 of Ova against the main protease (Mpro) of the SARS-CoV-2 virus is 7.316 µM. Our in vitro studies and the use of the periodontal ligament organoid model highlight Ovatodiolide's potential as a small molecule therapeutic agent that impedes the virus's ability to bind to the Neuropilin-1 receptor on host cells. The research uncovers various pathways and biochemical strategies through which Ovatodiolide may function as an effective antiviral small molecule drug.


Subject(s)
COVID-19 Drug Treatment , Neuropilin-1 , Organoids , Periodontal Ligament , SARS-CoV-2 , Periodontal Ligament/metabolism , Periodontal Ligament/cytology , Periodontal Ligament/virology , Humans , Organoids/virology , Organoids/metabolism , Organoids/drug effects , Neuropilin-1/metabolism , SARS-CoV-2/drug effects , Antiviral Agents/pharmacology , COVID-19/metabolism , COVID-19/virology , Diterpenes/pharmacology
15.
Aging (Albany NY) ; 16(9): 8086-8109, 2024 05 08.
Article in English | MEDLINE | ID: mdl-38728245

ABSTRACT

BACKGROUND: Research has shown a connection between vasculogenic mimicry (VM) and cancer progression. However, the functions of genes related to VM in the emergence and progression of TNBC have not been completely elucidated. METHODS: A survival risk model was constructed by screening biomarkers using DESeq2 and WGCNA based on public TNBC transcriptome data. Furthermore, gene set enrichment analysis was performed, and tumor microenvironment and drug sensitivity were analyzed. The selected biomarkers were validated via quantitative PCR detection, immunohistochemical staining, and protein detection in breast cancer cell lines. Biomarkers related to the proliferation and migration of TNBC cells were validated via in vitro experiments. RESULTS: The findings revealed that 235 target genes were connected to the complement and coagulation cascade pathways. The risk score was constructed using KCND2, NRP1, and VSTM4. The prognosis model using the risk score and pathological T stage yielded good validation results. The clinical risk of TNBC was associated with the angiogenesis signaling pathway, and the low-risk group exhibited better sensitivity to immunotherapy. Quantitative PCR and immunohistochemistry indicated that the expression levels of KCND2 in TNBC tissues were higher than those in adjacent nontumor tissues. In the TNBC cell line, the protein expression of KCND2 was increased. Knockdown of KCND2 and VSTM4 inhibited the proliferation and migration of TNBC cells in vitro. CONCLUSIONS: In this study, three VM-related biomarkers were identified, including KCND2, NRP1, and VSTM4. These findings are likely to aid in deepening our understanding of the regulatory mechanism of VM in TNBC.


Subject(s)
Biomarkers, Tumor , Neovascularization, Pathologic , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/metabolism , Female , Prognosis , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Tumor Microenvironment/genetics , Cell Proliferation/genetics , Neuropilin-1/genetics , Neuropilin-1/metabolism , Cell Movement/genetics , Transcriptome , Potassium Channels, Tandem Pore Domain/genetics , Potassium Channels, Tandem Pore Domain/metabolism
16.
Commun Biol ; 7(1): 629, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38789481

ABSTRACT

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.


Subject(s)
Endosomes , Endothelial Cells , Fibronectins , Integrin alpha5 , Neuropilin-1 , Neuropilin-2 , Proteomics , p120 GTPase Activating Protein , Animals , Mice , Endosomes/metabolism , Endothelial Cells/metabolism , Fibronectins/metabolism , Integrin alpha5/metabolism , Integrin alpha5/genetics , Integrins , Neuropilin-1/metabolism , Neuropilin-1/genetics , Neuropilin-2/metabolism , Neuropilin-2/genetics , p120 GTPase Activating Protein/metabolism , p120 GTPase Activating Protein/genetics , Protein Transport , Proteomics/methods
17.
Cell Host Microbe ; 32(6): 980-995.e9, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38729153

ABSTRACT

Mammalian orthoreovirus (reovirus) is a nonenveloped virus that establishes primary infection in the intestine and disseminates to sites of secondary infection, including the CNS. Reovirus entry involves multiple engagement factors, but how the virus disseminates systemically and targets neurons remains unclear. In this study, we identified murine neuropilin 1 (mNRP1) as a receptor for reovirus. mNRP1 binds reovirus with nanomolar affinity using a unique mechanism of virus-receptor interaction, which is coordinated by multiple interactions between distinct reovirus capsid subunits and multiple NRP1 extracellular domains. By exchanging essential capsid protein-encoding gene segments, we determined that the multivalent interaction is mediated by outer-capsid protein σ3 and capsid turret protein λ2. Using capsid mutants incapable of binding NRP1, we found that NRP1 contributes to reovirus dissemination and neurovirulence in mice. Collectively, our results demonstrate that NRP1 is an entry receptor for reovirus and uncover mechanisms by which NRPs promote viral entry and pathogenesis.


Subject(s)
Capsid Proteins , Neuropilin-1 , Orthoreovirus, Mammalian , Receptors, Virus , Reoviridae Infections , Virus Internalization , Animals , Mice , Capsid Proteins/metabolism , Capsid Proteins/genetics , Neuropilin-1/metabolism , Neuropilin-1/genetics , Orthoreovirus, Mammalian/genetics , Orthoreovirus, Mammalian/physiology , Orthoreovirus, Mammalian/metabolism , Reoviridae Infections/virology , Reoviridae Infections/metabolism , Receptors, Virus/metabolism , Humans , Capsid/metabolism , Cell Line , HEK293 Cells , Protein Binding , Mice, Inbred C57BL
18.
Int J Mol Sci ; 25(10)2024 May 16.
Article in English | MEDLINE | ID: mdl-38791476

ABSTRACT

Critical illness and sepsis may cause organ failure and are recognized as mortality drivers in hospitalized patients. Neuropilin-1 (NRP-1) is a multifaceted transmembrane protein involved in the primary immune response and is expressed in immune cells such as T and dendritic cells. The soluble form of NRP-1 (sNRP-1) acts as an antagonist to NRP-1 by scavenging its ligands. The aim of this study was to determine the value of sNRP-1 as a biomarker in critical illness and sepsis. We enrolled 180 critically ill patients admitted to a medical intensive care unit and measured serum sNRP-1 concentrations at admission, comparing them to 48 healthy individuals. Critically ill and septic patients showed higher levels of sNRP-1 compared to healthy controls (median of 2.47 vs. 1.70 nmol/L, p < 0.001). Moreover, sNRP-1 was also elevated in patients with sepsis compared to other critical illness (2.60 vs. 2.13 nmol/L, p = 0.01), irrespective of disease severity or organ failure. In critically ill patients, sNRP-1 is positively correlated with markers of kidney and hepatic dysfunction. Most notably, critically ill patients not surviving in the long term (one year after admission) showed higher concentrations of sNRP-1 at the time of ICU admission (p = 0.036), with this association being dependent on the presence of organ failure. Critically ill and septic patients exhibit higher serum concentrations of circulating sNRP-1, which correlates to organ failure, particularly hepatic and kidney dysfunction.


Subject(s)
Biomarkers , Critical Illness , Neuropilin-1 , Sepsis , Adult , Aged , Female , Humans , Male , Middle Aged , Biomarkers/blood , Case-Control Studies , Intensive Care Units , Multiple Organ Failure/blood , Multiple Organ Failure/etiology , Multiple Organ Failure/mortality , Neuropilin-1/metabolism , Neuropilin-1/blood , Sepsis/blood , Sepsis/mortality
19.
Biomed Pharmacother ; 176: 116766, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38788599

ABSTRACT

Activation of neuropilin-1 (NRP-1) by platelet derived growth factor (PDGF)-C sustains melanoma invasiveness. Therefore, in the search of novel agents capable of reducing melanoma spreading, PDGF-C/NRP-1 interaction was investigated as a potential druggable target. Since the PDGF-C region involved in NRP-1 binding is not yet known, based on the sequence and structural homology between PDGF-C and vascular endothelial growth factor-A (VEGF-A), we hypothesized that the NRP-1 b1 domain region involved in the interaction with VEGF-A might also be required for PDGF-C binding. Hence, this region was selected from the protein crystal structure and used as target in the molecular docking procedure. In the following virtual screening, compounds from a DrugBank database were used as query ligands to identify agents potentially capable of disrupting NRP-1/PDGF-C interaction. Among the top 45 candidates with the highest affinity, five drugs were selected based on the safety profile, lack of hormonal effects, and current availability in the market: the antipsychotic pimozide, antidiabetic gliclazide, antiallergic cromolyn sodium, anticancer tyrosine kinase inhibitor entrectinib, and antihistamine azelastine. Analysis of drug influence on PDGF-C in vitro binding to NRP-1 and PDGF-C induced migration of human melanoma cells expressing NRP-1, indicated gliclazide and entrectinib as the most specific agents that were active at clinically achievable and non-toxic concentrations. Both drugs also reverted PDGF-C ability to stimulate extracellular matrix invasion by melanoma cells resistant to BRAF inhibitors. The inhibitory effect on tumor cell motility involved a decrease of p130Cas phosphorylation, a signal transduction pathway activated by PDGF-C-mediated stimulation of NRP-1.


Subject(s)
Lymphokines , Melanoma , Molecular Docking Simulation , Neuropilin-1 , Platelet-Derived Growth Factor , Humans , Melanoma/drug therapy , Melanoma/pathology , Melanoma/metabolism , Lymphokines/metabolism , Platelet-Derived Growth Factor/metabolism , Neuropilin-1/metabolism , Cell Line, Tumor , Protein Binding , Cell Movement/drug effects , Neoplasm Metastasis , Antineoplastic Agents/pharmacology
20.
J Cell Mol Med ; 28(8): e18201, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38568078

ABSTRACT

Sensory nerves play a crucial role in maintaining bone homeostasis by releasing Semaphorin 3A (Sema3A). However, the specific mechanism of Sema3A in regulation of bone marrow mesenchymal stem cells (BMMSCs) during bone remodelling remains unclear. The tibial denervation model was used and the denervated tibia exhibited significantly lower mass as compared to sham operated bones. In vitro, BMMSCs cocultured with dorsal root ganglion cells (DRGs) or stimulated by Sema3A could promote osteogenic differentiation through the Wnt/ß-catenin/Nrp1 positive feedback loop, and the enhancement of osteogenic activity could be inhibited by SM345431 (Sema3A-specific inhibitor). In addition, Sema3A-stimulated BMMSCs or intravenous injection of Sema3A could promote new bone formation in vivo. To sum up, the coregulation of bone remodelling is due to the ageing of BMMSCs and increased osteoclast activity. Furthermore, the sensory neurotransmitter Sema3A promotes osteogenic differentiation of BMMSCs via Wnt/ß-catenin/Nrp1 positive feedback loop, thus promoting osteogenesis in vivo and in vitro.


Subject(s)
Mesenchymal Stem Cells , Osteogenesis , Osteogenesis/genetics , Semaphorin-3A/genetics , Feedback , beta Catenin , Ganglia, Spinal , Neuropilin-1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL