1
|
Grebinoski S, Pieklo G, Zhang Q, Visperas A, Cui J, Goulet J, Xiao H, Brunazzi EA, Cardello C, Herrada AA, Das J, Workman CJ, Vignali DAA. Regulatory T Cell Insufficiency in Autoimmune Diabetes Is Driven by Selective Loss of Neuropilin-1 on Intraislet Regulatory T Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:779-794. [PMID: 39109924 PMCID: PMC11371503 DOI: 10.4049/jimmunol.2300216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 07/08/2024] [Indexed: 09/05/2024]
Abstract
Approaches to reverse or limit regulatory T cell (Treg) insufficiency are of great interest for development of immunotherapeutic treatments for autoimmune patients, including type 1 diabetes. Treg insufficiency is heavily implicated in the progression of autoimmune diabetes in the NOD mouse model and is characterized by defects in Treg numbers, development, and/or function. Utilizing a Treg-centric screen, we show that intraislet Tregs have a uniquely dysfunctional phenotype, hallmarked by an almost complete lack of neuropilin-1 (Nrp1), a cell surface receptor required to maintain Treg stability. Intraislet Nrp1- Tregs exhibit hallmark features of fragility, including reduced suppressive capacity, decreased CD73 and Helios, and increased Rorγt and Tbet. Intraislet Nrp1- Tregs also exhibit decreased Foxp3 expression on a per cell basis, suggesting that Nrp1 may also be required for long-term Treg stability. Mechanistically, Treg-restricted augmentation of Nrp1 expression limited the onset of autoimmune diabetes in NOD mice suggesting that Nrp1 critically impacts intraislet Treg function. Transcriptional analysis showed that Nrp1 restoration led to an increase in markers and pathways of TCR signaling, survival, and suppression, and when Nrp1 protein expression is examined by cellular indexing of transcriptomes and epitopes by sequencing, significant differences were observed between Nrp1+ and Nrp1- Tregs in all tissues, particularly in markers of Treg fragility. This translated into substantive differences between Nrp1+ and Nrp1- Tregs that afforded the former with a competitive advantage in the islets. Taken together, these data suggest that maintenance of Nrp1 expression and signaling on Tregs limits diabetes onset and may serve as a strategy to combat Treg insufficiency in autoimmune disease.
Collapse
Affiliation(s)
- Stephanie Grebinoski
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Graduate Program of Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA
| | - Gwenyth Pieklo
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA
| | - Qianxia Zhang
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Graduate Program of Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN
| | - Anabelle Visperas
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA
| | - Jian Cui
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA
| | - Jordana Goulet
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Hanxi Xiao
- Center for Systems Immunology, Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Computational & Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- CMU-Pitt Joint Computational Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Erin A Brunazzi
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA
| | - Carly Cardello
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA
| | - Andrés A Herrada
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN
| | - Jishnu Das
- CMU-Pitt Joint Computational Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Creg J Workman
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh PA
| |
Collapse
|
2
|
Matilla L, Martín-Núñez E, Navarro A, Garaikoetxea M, Fernández-Celis A, Goñi-Olóriz M, Gainza A, Fernández-Irigoyen J, Santamaría E, Tamayo I, Álvarez V, Sádaba R, Jover E, López-Andrés N. Neuropilin-1 sex-dependently modulates inflammatory, angiogenic and osteogenic phenotypes in the calcifying valve interstitial cell. Biochem Pharmacol 2024; 226:116336. [PMID: 38844264 DOI: 10.1016/j.bcp.2024.116336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 05/29/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024]
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.
Collapse
Affiliation(s)
- Lara Matilla
- Cardiovascular Translational Research, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, IdiSNA, Pamplona, Spain
| | - Ernesto Martín-Núñez
- Cardiovascular Translational Research, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, IdiSNA, Pamplona, Spain
| | - Adela Navarro
- Cardiovascular Translational Research, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, IdiSNA, Pamplona, Spain
| | - Mattie Garaikoetxea
- Cardiovascular Translational Research, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, IdiSNA, Pamplona, Spain
| | - Amaya Fernández-Celis
- Cardiovascular Translational Research, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, IdiSNA, Pamplona, Spain
| | - Miriam Goñi-Olóriz
- Cardiovascular Translational Research, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, IdiSNA, Pamplona, Spain
| | - Alicia Gainza
- Cardiovascular Translational Research, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, IdiSNA, Pamplona, Spain
| | - Joaquín Fernández-Irigoyen
- Clinical Neuroproteomics Unit, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, IdiSNA, Pamplona, Spain
| | - Enrique Santamaría
- Clinical Neuroproteomics Unit, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, IdiSNA, Pamplona, Spain
| | - Ibai Tamayo
- Research Methodology Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain
| | - Virginia Álvarez
- Cardiovascular Translational Research, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, IdiSNA, Pamplona, Spain
| | - Rafael Sádaba
- Cardiovascular Translational Research, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, IdiSNA, Pamplona, Spain
| | - Eva Jover
- Cardiovascular Translational Research, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, IdiSNA, Pamplona, Spain.
| | - Natalia López-Andrés
- Cardiovascular Translational Research, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, IdiSNA, Pamplona, Spain.
| |
Collapse
|
3
|
Joyce JD, Moore GA, Goswami P, Harrell TL, Taylor TM, Hawks SA, Green JC, Jia M, Irwin MD, Leslie E, Duggal NK, Thompson CK, Bertke AS. SARS-CoV-2 Rapidly Infects Peripheral Sensory and Autonomic Neurons, Contributing to Central Nervous System Neuroinvasion before Viremia. Int J Mol Sci 2024; 25:8245. [PMID: 39125815 PMCID: PMC11311394 DOI: 10.3390/ijms25158245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
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.
Collapse
Affiliation(s)
- Jonathan D. Joyce
- Translational Biology, Medicine, and Health, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA; (J.D.J.)
- Center for Emerging Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
| | - Greyson A. Moore
- Biomedical and Veterinary Science, Virginia Maryland College of Veterinary Medicine, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
| | - Poorna Goswami
- Translational Biology, Medicine, and Health, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA; (J.D.J.)
| | - Telvin L. Harrell
- Biomedical and Veterinary Science, Virginia Maryland College of Veterinary Medicine, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
| | - Tina M. Taylor
- Population Health Sciences, Virginia Maryland College of Veterinary Medicine, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
| | - Seth A. Hawks
- Biomedical Sciences and Pathobiology, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
| | - Jillian C. Green
- Biomedical and Veterinary Science, Virginia Maryland College of Veterinary Medicine, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
| | - Mo Jia
- Population Health Sciences, Virginia Maryland College of Veterinary Medicine, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
| | - Matthew D. Irwin
- Biomedical and Veterinary Science, Virginia Maryland College of Veterinary Medicine, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
| | - Emma Leslie
- Translational Biology, Medicine, and Health, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA; (J.D.J.)
| | - Nisha K. Duggal
- Center for Emerging Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
- Biomedical Sciences and Pathobiology, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
| | - Christopher K. Thompson
- School of Neuroscience, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
| | - Andrea S. Bertke
- Center for Emerging Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
- Population Health Sciences, Virginia Maryland College of Veterinary Medicine, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
| |
Collapse
|
4
|
Guinn S, Kinny-Köster B, Tandurella JA, Mitchell JT, Sidiropoulos DN, Loth M, Lyman MR, Pucsek AB, Zabransky DJ, Lee JW, Kartalia E, Ramani M, Seppälä TT, Cherry C, Suri R, Zlomke H, Patel J, He J, Wolfgang CL, Yu J, Zheng L, Ryan DP, Ting DT, Kimmelman A, Gupta A, Danilova L, Elisseeff JH, Wood LD, Stein-O’Brien G, Kagohara LT, Jaffee EM, Burkhart RA, Fertig EJ, Zimmerman JW. Transfer Learning Reveals Cancer-Associated Fibroblasts Are Associated with Epithelial-Mesenchymal Transition and Inflammation in Cancer Cells in Pancreatic Ductal Adenocarcinoma. Cancer Res 2024; 84:1517-1533. [PMID: 38587552 PMCID: PMC11065624 DOI: 10.1158/0008-5472.can-23-1660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/09/2023] [Accepted: 10/27/2023] [Indexed: 04/09/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy characterized by an immunosuppressive tumor microenvironment enriched with cancer-associated fibroblasts (CAF). This study used a convergence approach to identify tumor cell and CAF interactions through the integration of single-cell data from human tumors with human organoid coculture experiments. Analysis of a comprehensive atlas of PDAC single-cell RNA sequencing data indicated that CAF density is associated with increased inflammation and epithelial-mesenchymal transition (EMT) in epithelial cells. Transfer learning using transcriptional data from patient-derived organoid and CAF cocultures provided in silico validation of CAF induction of inflammatory and EMT epithelial cell states. Further experimental validation in cocultures demonstrated integrin beta 1 (ITGB1) and vascular endothelial factor A (VEGFA) interactions with neuropilin-1 mediating CAF-epithelial cell cross-talk. Together, this study introduces transfer learning from human single-cell data to organoid coculture analyses for experimental validation of discoveries of cell-cell cross-talk and identifies fibroblast-mediated regulation of EMT and inflammation. SIGNIFICANCE Adaptation of transfer learning to relate human single-cell RNA sequencing data to organoid-CAF cocultures facilitates discovery of human pancreatic cancer intercellular interactions and uncovers cross-talk between CAFs and tumor cells through VEGFA and ITGB1.
Collapse
Affiliation(s)
- Samantha Guinn
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Benedict Kinny-Köster
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Surgery, New York University Grossman School of Medicine, New York, NY
| | - Joseph A. Tandurella
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jacob T. Mitchell
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD
| | - Dimitrios N. Sidiropoulos
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Melanie Loth
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Melissa R. Lyman
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Alexandra B. Pucsek
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Daniel J. Zabransky
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jae W. Lee
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD
| | - Emma Kartalia
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Mili Ramani
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Toni T. Seppälä
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
- Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere University Hospital
| | - Christopher Cherry
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD
- Translational Tissue Engineering Center, Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD
| | - Reecha Suri
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Haley Zlomke
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jignasha Patel
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jin He
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | - Jun Yu
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Lei Zheng
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD
| | - David P. Ryan
- The Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - David T. Ting
- The Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Alec Kimmelman
- Department of Radiation Oncology at New York University Grossman School of Medicine, NYU Langone Health, New York, New York
| | - Anuj Gupta
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ludmila Danilova
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jennifer H. Elisseeff
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD
- Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere University Hospital
- Translational Tissue Engineering Center, Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD
| | - Laura D. Wood
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD
| | - Genevieve Stein-O’Brien
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD
| | - Luciane T. Kagohara
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Elizabeth M. Jaffee
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Richard A. Burkhart
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD
| | - Elana J. Fertig
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD
- Department of Applied Mathematics and Statistics, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD
| | - Jacquelyn W. Zimmerman
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
5
|
Lechner K, Kia S, von Korn P, Dinges SM, Mueller S, Tjønna AE, Wisløff U, Van Craenenbroeck EM, Pieske B, Adams V, Pressler A, Landmesser U, Halle M, Kränkel N. Cardiometabolic and immune response to exercise training in patients with metabolic syndrome: retrospective analysis of two randomized clinical trials. Front Cardiovasc Med 2024; 11:1329633. [PMID: 38638882 PMCID: PMC11025358 DOI: 10.3389/fcvm.2024.1329633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 03/21/2024] [Indexed: 04/20/2024] Open
Abstract
Background Metabolic syndrome (MetS) is defined by the presence of central obesity plus ≥two metabolic/cardiovascular risk factors (RF), with inflammation being a major disease-driving mechanism. Structured endurance exercise training (ET) may positively affect these traits, as well as cardiorespiratory fitness (V̇O2peak). Aims We explore individual ET-mediated improvements of MetS-associated RF in relation to improvements in V̇O2peak and inflammatory profile. Methods MetS patients from two randomized controlled trials, ExMET (n = 24) and OptimEx (n = 34), had performed 4- or 3-months supervised ET programs according to the respective trial protocol. V̇O2peak, MetS-defining RFs (both RCTs), broad blood leukocyte profile, cytokines and plasma proteins (ExMET only) were assessed at baseline and follow-up. Intra-individual changes in RFs were analysed for both trials separately using non-parametric approaches. Associations between changes in each RF over the exercise period (n-fold of baseline values) were correlated using a non-parametrical approach (Spearman). RF clustering was explored by uniform manifold approximation and projection (UMAP) and changes in RF depending on other RF or exercise parameters were explored by recursive partitioning. Results Four months of ET reduced circulating leukocyte counts (63.5% of baseline, P = 8.0e-6), especially effector subtypes. ET response of MetS-associated RFs differed depending on patients' individual RF constellation, but was not associated with individual change in V̇O2peak. Blood pressure lowering depended on cumulative exercise duration (ExMET: ≥102 min per week; OptimEx-MetS: ≥38 min per session) and baseline triglyceride levels (ExMET: <150 mg/dl; OptimEx-MetS: <174.8 mg/dl). Neuropilin-1 plasma levels were inversely associated with fasting plasma triglycerides (R: -0.4, P = 0.004) and changes of both parameters during the ET phase were inversely correlated (R: -0.7, P = 0.0001). Conclusions ET significantly lowered effector leukocyte blood counts. The improvement of MetS-associated cardiovascular RFs depended on individual basal RF profile and exercise duration but was not associated with exercise-mediated increase in V̇O2peak. Neuropilin-1 may be linked to exercise-mediated triglyceride lowering.
Collapse
Affiliation(s)
- Katharina Lechner
- Department of Prevention and Sports Medicine, University Hospital Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- DZHK, German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
| | - Sylvia Kia
- Deutsches Herzzentrum der Charité, Klinik für Kardiologie, Angiologie und Intensivmedizin, Berlin, Germany
- DZHK, German Centre for Cardiovascular Research, Partner Site, Berlin, Germany
| | - Pia von Korn
- Department of Prevention and Sports Medicine, University Hospital Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- DZHK, German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Sophia M. Dinges
- Department of Prevention and Sports Medicine, University Hospital Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- DZHK, German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Stephan Mueller
- Department of Prevention and Sports Medicine, University Hospital Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- DZHK, German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Arnt-Erik Tjønna
- Cardiac Exercise Research Group (CERG), Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ulrik Wisløff
- Cardiac Exercise Research Group (CERG), Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
- Centre for Research on Exercise, Physical Activity and Health, School of Human Movement and Nutrition Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Emeline M. Van Craenenbroeck
- Research Group Cardiovascular Diseases, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital (UZA), Edegem, Belgium
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Campus Virchow Klinikum, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Volker Adams
- Department of Cardiology and Internal Medicine, Heart Center Dresden-University Hospital, TU Dresden, Dresden, Germany
| | - Axel Pressler
- Department of Prevention and Sports Medicine, University Hospital Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- DZHK, German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
- Private Center for Sports and Exercise Cardiology, Munich, Germany
| | - Ulf Landmesser
- Deutsches Herzzentrum der Charité, Klinik für Kardiologie, Angiologie und Intensivmedizin, Berlin, Germany
- DZHK, German Centre for Cardiovascular Research, Partner Site, Berlin, Germany
- Friede Springer—Centre of Cardiovascular Prevention at Charité, Charité University Medicine Berlin, Berlin, Germany
| | - Martin Halle
- Department of Prevention and Sports Medicine, University Hospital Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- DZHK, German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Nicolle Kränkel
- Deutsches Herzzentrum der Charité, Klinik für Kardiologie, Angiologie und Intensivmedizin, Berlin, Germany
- DZHK, German Centre for Cardiovascular Research, Partner Site, Berlin, Germany
- Friede Springer—Centre of Cardiovascular Prevention at Charité, Charité University Medicine Berlin, Berlin, Germany
| |
Collapse
|
6
|
Pang PP, Zhang HY, Zhang DC, Tang JX, Gong Y, Guo YC, Zheng CB. Investigating the impact of protein S-sulfhydration modification on vascular diseases: A comprehensive review. Eur J Pharmacol 2024; 966:176345. [PMID: 38244760 DOI: 10.1016/j.ejphar.2024.176345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 01/22/2024]
Abstract
The post-translational modification of cysteine through redox reactions, especially S-sulfhydration, plays a critical role in regulating protein activity, interactions, and spatial arrangement. This review focuses on the impact of protein S-sulfhydration on vascular function and its implications in vascular diseases. Dysregulated S-sulfhydration has been linked to the development of vascular pathologies, including aortic aneurysms and dissections, atherosclerosis, and thrombotic diseases. The H2S signaling pathway and the enzyme cystathionine γ-lyase (CSE), which is responsible for H2S generation, are identified as key regulators of vascular function. Additionally, potential therapeutic targets for the treatment of vascular diseases, such as the H2S donor GYY4137 and the HDAC inhibitor entinostat, are discussed. The review also emphasizes the antithrombotic effects of H2S in regulating platelet aggregation and thrombosis. The aim of this review is to enhance our understanding of the function and mechanism of protein S-sulfhydration modification in vascular diseases, and to provide new insights into the clinical application of this modification.
Collapse
Affiliation(s)
- Pan-Pan Pang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, China
| | - Hong-Ye Zhang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, China
| | - Ding-Cheng Zhang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, China
| | - Jia-Xiang Tang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, China
| | - Yu Gong
- Yunnan Provincial Hospital of Infection Disease/ Yunnan AIDS Care Center/ Yunnan Mental Health Center, Kunming, 650301, China
| | - Yu-Chen Guo
- University of Sydney Pharmacy School, Sydney, 2006, Australia
| | - Chang-Bo Zheng
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, China; College of Modern Biomedical Industry, Kunming Medical University, Kunming, 650500, China; Yunnan Vaccine Laboratory, Kunming, 650500, China.
| |
Collapse
|
7
|
de Gea P, Benkeder S, Bouvet P, Aimard M, Chounlamountri N, Honnorat J, Do LD, Meissirel C. VEGF controls microglial phagocytic response to amyloid-β. Front Cell Neurosci 2023; 17:1264402. [PMID: 38162003 PMCID: PMC10757340 DOI: 10.3389/fncel.2023.1264402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 11/23/2023] [Indexed: 01/03/2024] Open
Abstract
Microglial cells are well known to be implicated in the pathogenesis of Alzheimer's disease (AD), due to the impaired clearance of amyloid-β (Aβ) protein. In AD, Aβ accumulates in the brain parenchyma as soluble oligomers and protofibrils, and its aggregation process further give rise to amyloid plaques. Compelling evidence now indicate that Aβ oligomers (Aβo) are the most toxic forms responsible for neuronal and synaptic alterations. Recently, we showed that the Vascular Endothelial Growth Factor (VEGF) counteracts Aβo-induced synaptic alterations and that a peptide derived from VEGF is able to inhibit Aβ aggregation process. Moreover, VEGF has been reported to promote microglial chemotaxis to Aβ brain deposits. We therefore investigated whether VEGF could influence microglial phagocytic response to Aβ, using in vitro and ex vivo models of amyloid accumulation. We report here that VEGF increases Aβo phagocytosis by microglial cells and further characterized the molecular basis of the VEGF effect. VEGF is able to control α-secretase activity in microglial cells, resulting in the increased cleavage of the Triggering Receptor Expressed on Myeloid cells 2 (TREM2), a major microglial Aβ receptor. Consistently, the soluble form sTREM2 also increases Aβo phagocytosis by microglial cells. Taken together, these findings propose VEGF as a new regulator of Aβ clearance and suggest its potential role in rescuing compromised microglial function in AD.
Collapse
Affiliation(s)
- Priscille de Gea
- Laboratory MeLIS, Institut Neuromyogène, Synaptopathies and Autoantibodies, INSERM U1314, CNRS UMR 5284, Université Claude Bernard Lyon 1, Lyon, France
| | - Sarah Benkeder
- Laboratory MeLIS, Institut Neuromyogène, Synaptopathies and Autoantibodies, INSERM U1314, CNRS UMR 5284, Université Claude Bernard Lyon 1, Lyon, France
| | - Pauline Bouvet
- Laboratory MeLIS, Institut Neuromyogène, Synaptopathies and Autoantibodies, INSERM U1314, CNRS UMR 5284, Université Claude Bernard Lyon 1, Lyon, France
| | - Mélanie Aimard
- Laboratory MeLIS, Institut Neuromyogène, Synaptopathies and Autoantibodies, INSERM U1314, CNRS UMR 5284, Université Claude Bernard Lyon 1, Lyon, France
| | - Naura Chounlamountri
- Laboratory MeLIS, Institut Neuromyogène, Synaptopathies and Autoantibodies, INSERM U1314, CNRS UMR 5284, Université Claude Bernard Lyon 1, Lyon, France
| | - Jérôme Honnorat
- Laboratory MeLIS, Institut Neuromyogène, Synaptopathies and Autoantibodies, INSERM U1314, CNRS UMR 5284, Université Claude Bernard Lyon 1, Lyon, France
- French Reference Center for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, Hôpital Neurologique Pierre Wertheimer, Bron, France
| | - Le Duy Do
- Laboratory MeLIS, Institut Neuromyogène, Synaptopathies and Autoantibodies, INSERM U1314, CNRS UMR 5284, Université Claude Bernard Lyon 1, Lyon, France
- French Reference Center for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, Hôpital Neurologique Pierre Wertheimer, Bron, France
| | - Claire Meissirel
- Laboratory MeLIS, Institut Neuromyogène, Synaptopathies and Autoantibodies, INSERM U1314, CNRS UMR 5284, Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
8
|
Ngaha TYS, Zhilenkova AV, Essogmo FE, Uchendu IK, Abah MO, Fossa LT, Sangadzhieva ZD, D. Sanikovich V, S. Rusanov A, N. Pirogova Y, Boroda A, Rozhkov A, Kemfang Ngowa JD, N. Bagmet L, I. Sekacheva M. Angiogenesis in Lung Cancer: Understanding the Roles of Growth Factors. Cancers (Basel) 2023; 15:4648. [PMID: 37760616 PMCID: PMC10526378 DOI: 10.3390/cancers15184648] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/11/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Research has shown the role of growth factors in lung cancer angiogenesis. Angiogenesis promotes lung cancer progression by stimulating tumor growth, enhancing tumor invasion, contributing to metastasis, and modifying immune system responses within the tumor microenvironment. As a result, new treatment techniques based on the anti-angiogenic characteristics of compounds have been developed. These compounds selectively block the growth factors themselves, their receptors, or the downstream signaling pathways activated by these growth factors. The EGF and VEGF families are the primary targets in this approach, and several studies are being conducted to propose anti-angiogenic drugs that are increasingly suitable for the treatment of lung cancer, either as monotherapy or as combined therapy. The efficacy of the results are encouraging, but caution must be placed on the higher risk of toxicity, outlining the importance of personalized follow-up in the management of these patients.
Collapse
Affiliation(s)
- Tchawe Yvan Sinclair Ngaha
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
- Department of Public Health, James Lind Institute, Rue de la Cité 1, 1204 Geneva, Switzerland
| | - Angelina V. Zhilenkova
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
| | - Freddy Elad Essogmo
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
| | - Ikenna K. Uchendu
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
- Medical Laboratory Science Department, Faculty of Health Science and Technology, College of Medicine, University of Nigeria, Enugu Campus, Enugu 410001, Nigeria
| | - Moses Owoicho Abah
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
| | - Lionel Tabola Fossa
- Department of Oncology, Bafoussam Regional Hospital, Bafoussam 980, Cameroon;
| | - Zaiana D. Sangadzhieva
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
| | - Varvara D. Sanikovich
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
| | - Alexander S. Rusanov
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
| | - Yuliya N. Pirogova
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
| | - Alexander Boroda
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
| | - Alexander Rozhkov
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
| | - Jean D. Kemfang Ngowa
- Faculty of Medicine and Biomedical Sciences, University of Yaounde I, Yaounde 1364, Cameroon;
| | - Leonid N. Bagmet
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
| | - Marina I. Sekacheva
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
| |
Collapse
|
9
|
Izci M, Maksoudian C, Gonçalves F, Pérez Gilabert I, Rios Luci C, Bolea-Fernandez E, Vanhaecke F, Manshian BB, Soenen SJ. The Efficacy of Nanoparticle Delivery to Hypoxic Solid Tumors by ciRGD Co-Administration Depends on Neuropilin-1 and Neutrophil Levels. Adv Healthc Mater 2023; 12:e2300594. [PMID: 37247322 DOI: 10.1002/adhm.202300594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/02/2023] [Indexed: 05/31/2023]
Abstract
The ability to improve nanoparticle delivery to solid tumors is an actively studied domain, where various mechanisms are looked into. In previous work, the authors have looked into nanoparticle size, tumor vessel normalization, and disintegration, and here it is aimed to continue this work by performing an in-depth mechanistic study on the use of ciRGD peptide co-administration. Using a multiparametric approach, it is observed that ciRGD can improve nanoparticle delivery to the tumor itself, but also to tumor cells specifically better than vessel normalization strategies. The effect depends on the level of tumor perfusion, hypoxia, neutrophil levels, and vessel permeability. This work shows that upon characterizing tumors for these parameters, conditions can be selected that can optimally benefit from ciRGD co-administration as a means to improve NP delivery to solid tumors.
Collapse
Affiliation(s)
- Mukaddes Izci
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven Herestraat 49, Leuven, B3000, Belgium
| | - Christy Maksoudian
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven Herestraat 49, Leuven, B3000, Belgium
| | - Filipa Gonçalves
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven Herestraat 49, Leuven, B3000, Belgium
| | - Irati Pérez Gilabert
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven Herestraat 49, Leuven, B3000, Belgium
| | - Carla Rios Luci
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven Herestraat 49, Leuven, B3000, Belgium
| | - Eduardo Bolea-Fernandez
- Atomic & Mass Spectrometry - A&MS research group, Department of Chemistry, Ghent University, Campus Sterre, Krijgslaan 281-S12, Ghent, 9000, Belgium
| | - Frank Vanhaecke
- Atomic & Mass Spectrometry - A&MS research group, Department of Chemistry, Ghent University, Campus Sterre, Krijgslaan 281-S12, Ghent, 9000, Belgium
| | - Bella B Manshian
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven Herestraat 49, Leuven, B3000, Belgium
- Leuven Cancer Research Institute, Faculty of Medical Sciences, KU Leuven, Herestraat 49, Leuven, B3000, Belgium
| | - Stefaan J Soenen
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven Herestraat 49, Leuven, B3000, Belgium
- Leuven Cancer Research Institute, Faculty of Medical Sciences, KU Leuven, Herestraat 49, Leuven, B3000, Belgium
| |
Collapse
|
10
|
Potassium Dehydroandrograpolide Succinate Targets NRP1 Mediated VEGFR2/VE-Cadherin Signaling Pathway to Promote Endothelial Barrier Repair. Int J Mol Sci 2023; 24:ijms24043096. [PMID: 36834519 PMCID: PMC9964616 DOI: 10.3390/ijms24043096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/25/2023] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
Impairment of vascular endothelial integrity is associated with various vascular diseases. Our previous studies demonstrated that andrographolide is critical to maintaining gastric vascular homeostasis, as well as to regulating pathological vascular remodeling. Potassium dehydroandrograpolide succinate (PDA), a derivative of andrographolide, has been clinically used for the therapeutic treatment of inflammatory diseases. This study aimed to determine whether PDA promotes endothelial barrier repair in pathological vascular remodeling. Partial ligation of the carotid artery in ApoE-/- mice was used to evaluate whether PDA can regulate pathological vascular remodeling. A flow cytometry assay, BRDU incorporation assay, Boyden chamber cell migration assay, spheroid sprouting assay and Matrigel-based tube formation assay were performed to determine whether PDA can regulate the proliferation and motility of HUVEC. A molecular docking simulation and CO-immunoprecipitation assay were performed to observe protein interactions. We observed that PDA induced pathological vascular remodeling characterized by enhanced neointima formation. PDA treatment significantly enhanced the proliferation and migration of vascular endothelial cells. Investigating the potential mechanisms and signaling pathways, we observed that PDA induced endothelial NRP1 expression and activated the VEGF signaling pathway. Knockdown of NRP1 using siRNA transfection attenuated PDA-induced VEGFR2 expression. The interaction between NRP1 and VEGFR2 caused VE-Cad-dependent endothelial barrier impairment, which was characterized by enhanced vascular inflammation. Our study demonstrated that PDA plays a critical role in promoting endothelial barrier repair in pathological vascular remodeling.
Collapse
|
11
|
Al-Thomali AW, Al-kuraishy HM, Al-Gareeb AI, K. Al-buhadiliy A, De Waard M, Sabatier JM, Khan Khalil AA, Saad HM, Batiha GES. Role of Neuropilin 1 in COVID-19 Patients with Acute Ischemic Stroke. Biomedicines 2022; 10:2032. [PMID: 36009579 PMCID: PMC9405641 DOI: 10.3390/biomedicines10082032] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/13/2022] [Accepted: 08/17/2022] [Indexed: 12/13/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) infection can trigger the adaptive and innate immune responses, leading to uncontrolled inflammatory reactions and associated local and systematic tissue damage, along with thromboembolic disorders that may increase the risk of acute ischemic stroke (AIS) in COVID-19 patients. The neuropilin (NRP-1) which is a co-receptor for the vascular endothelial growth factor (VEGF), integrins, and plexins, is involved in the pathogenesis of AIS. NRP-1 is also regarded as a co-receptor for the entry of SARS-CoV-2 and facilitates its entry into the brain through the olfactory epithelium. NRP-1 is regarded as a cofactor for binding of SARS-CoV-2 with angiotensin-converting enzyme 2 (ACE2), since the absence of ACE2 reduces SARS-CoV-2 infectivity even in presence of NRP-1. Therefore, the aim of the present study was to clarify the potential role of NRP-1 in COVID-19 patients with AIS. SARS-CoV-2 may transmit to the brain through NRP-1 in the olfactory epithelium of the nasal cavity, leading to different neurological disorders, and therefore about 45% of COVID-19 patients had neurological manifestations. NRP-1 has the potential capability to attenuate neuroinflammation, blood-brain barrier (BBB) permeability, cerebral endothelial dysfunction (ED), and neuronal dysfunction that are uncommon in COVID-19 with neurological involvement, including AIS. Similarly, high NRP-1 serum level is linked with ED, oxidative stress, and the risk of pulmonary thrombosis in patients with severe COVID-19, suggesting a compensatory mechanism to overcome immuno-inflammatory disorders. In conclusion, NRP-1 has an important role in the pathogenesis of COVID-19 and AIS, and could be the potential biomarker linking the development of AIS in COVID-19. The present findings cannot provide a final conclusion, and thus in silico, experimental, in vitro, in vivo, preclinical, and clinical studies are recommended to confirm the potential role of NRP-1 in COVID-19, and to elucidate the pharmacological role of NRP-1 receptor agonists and antagonists in COVID-19.
Collapse
Affiliation(s)
- Asma W. Al-Thomali
- Department of Biology, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Hayder M. Al-kuraishy
- Department of Pharmacology, Toxicology and Medicine, Medical Faculty, College of Medicine, Al-Mustansiriyah University, MBChB, MRCP, FRCP, Baghdad P.O. Box 14132, Iraq
| | - Ali I. Al-Gareeb
- Department of Pharmacology, Toxicology and Medicine, Medical Faculty, College of Medicine, Al-Mustansiriyah University, Baghdad P.O. Box 14132, Iraq
| | - Ali K. Al-buhadiliy
- Department of Clinical Pharmacology, Medicine and Therapeutic, Medical Faculty, College of Medicine, Al-Mustansiriyah University, Baghdad P.O. Box 14132, Iraq
| | - Michel De Waard
- Smartox Biotechnology, 6 rue des Platanes, 38120 Saint-Egrève, France
- L’institut du Thorax, INSERM, CNRS, UNIV NANTES, 44007 Nantes, France
- LabEx «Ion Channels, Science & Therapeutics», Université de Nice Sophia-Antipolis, 06560 Valbonne, France
| | - Jean-Marc Sabatier
- Institut de Neurophysiopathologie (INP), Aix-Marseille Université, CNRS UMR 7051, Faculté des Sciences Médicales et Paramédicales, 27 Bd Jean Moulin, 13005 Marseille, France
| | - Atif Ali Khan Khalil
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi 46000, Pakistan
| | - Hebatallah M. Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Matrouh 51744, Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt
| |
Collapse
|
12
|
Gupta R, Sahu M, Tripathi R, Ambasta RK, Kumar P. Protein S-sulfhydration: Unraveling the prospective of hydrogen sulfide in the brain, vasculature and neurological manifestations. Ageing Res Rev 2022; 76:101579. [PMID: 35124235 DOI: 10.1016/j.arr.2022.101579] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 01/30/2022] [Accepted: 02/01/2022] [Indexed: 02/08/2023]
Abstract
Hydrogen sulfide (H2S) and hydrogen polysulfides (H2Sn) are essential regulatory signaling molecules generated by the entire body, including the central nervous system. Researchers have focused on the classical H2S signaling from the past several decades, whereas the last decade has shown the emergence of H2S-induced protein S-sulfhydration signaling as a potential therapeutic approach. Cysteine S-persulfidation is a critical paradigm of post-translational modification in the process of H2S signaling. Additionally, studies have shown the cross-relationship between S-sulfhydration and other cysteine-induced post-translational modifications, namely nitrosylation and carbonylation. In the central nervous system, S-sulfhydration is involved in the cytoprotection through various signaling pathways, viz. inflammatory response, oxidative stress, endoplasmic reticulum stress, atherosclerosis, thrombosis, and angiogenesis. Further, studies have demonstrated H2S-induced S-sulfhydration in regulating different biological processes, such as mitochondrial integrity, calcium homeostasis, blood-brain permeability, cerebral blood flow, and long-term potentiation. Thus, protein S-sulfhydration becomes a crucial regulatory molecule in cerebrovascular and neurodegenerative diseases. Herein, we first described the generation of intracellular H2S followed by the application of H2S in the regulation of cerebral blood flow and blood-brain permeability. Further, we described the involvement of S-sulfhydration in different biological and cellular functions, such as inflammatory response, mitochondrial integrity, calcium imbalance, and oxidative stress. Moreover, we highlighted the importance of S-sulfhydration in cerebrovascular and neurodegenerative diseases.
Collapse
|
13
|
Qiu J, Chen R, Zhao L, Lian C, Liu Z, Zhu X, Cui J, Wang S, Wang M, Huang Y, Wang S, Wang J. Circular RNA circGSE1 promotes angiogenesis in ageing mice by targeting the miR-323-5p/NRP1 axis. Aging (Albany NY) 2022; 14:3049-3069. [PMID: 35366240 PMCID: PMC9037273 DOI: 10.18632/aging.203988] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 02/15/2022] [Indexed: 11/29/2022]
Abstract
Age is an important factor in many cardiovascular diseases, in which endothelial cells (ECs) play an important role. Circular RNAs (circRNAs) have been reported in many cardiovascular diseases, but their role in ageing EC-related angiogenesis is unclear. We aimed to identify a functional circRNA that regulates angiogenesis during ageing and explore its specific mechanism. In this study, we searched for differentially expressed circRNAs in old endothelial cells (OECs) and young endothelial cells (YECs) by circRNA sequencing and found that circGSE1 was significantly downregulated in OECs. Our study showed that circGSE1 could promote the proliferation, migration and tube formation of OECs in vitro. In a mouse model of femoral artery ligation and ischemia, circGSE1 promoted blood flow recovery and angiogenesis in the ischemic limbs of ageing mice. Mechanistically, we found that overexpressing circGSE1 reduced miR-323-5p expression, increased neuropilin-1 (NRP1) expression, and promoted proliferation, migration, and tube formation in OECs, while knocking down circGSE1 increased miR-323-5p expression, reduced NRP1 expression, and inhibited proliferation, migration, and tube formation in YECs. During EC ageing, circGSE1 may act through the miR-323-5p/NRP1 axis and promote endothelial angiogenesis in mice. Finally, the circGSE1/miR-323-5p/NRP1 axis could serve as a potential and promising therapeutic target for angiogenesis during ageing.
Collapse
Affiliation(s)
- Jiacong Qiu
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, China.,National-Local Joint Engineering Laboratory of Vascular Disease Treatment, Guangzhou 510080, Guangdong, China.,Guangdong Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou 510080, Guangdong, China
| | - Rencong Chen
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, China.,National-Local Joint Engineering Laboratory of Vascular Disease Treatment, Guangzhou 510080, Guangdong, China.,Guangdong Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou 510080, Guangdong, China
| | - Lei Zhao
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, China.,National-Local Joint Engineering Laboratory of Vascular Disease Treatment, Guangzhou 510080, Guangdong, China.,Guangdong Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou 510080, Guangdong, China
| | - Chong Lian
- Department of Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, Henan, China
| | - Zhen Liu
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, China.,National-Local Joint Engineering Laboratory of Vascular Disease Treatment, Guangzhou 510080, Guangdong, China.,Guangdong Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou 510080, Guangdong, China
| | - Xiaonan Zhu
- Department of Pharmacology Laboratory, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, Guangdong, China
| | - Jin Cui
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, China.,National-Local Joint Engineering Laboratory of Vascular Disease Treatment, Guangzhou 510080, Guangdong, China.,Guangdong Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou 510080, Guangdong, China
| | - Siwen Wang
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, China.,National-Local Joint Engineering Laboratory of Vascular Disease Treatment, Guangzhou 510080, Guangdong, China.,Guangdong Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou 510080, Guangdong, China
| | - Mingshan Wang
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, China.,National-Local Joint Engineering Laboratory of Vascular Disease Treatment, Guangzhou 510080, Guangdong, China.,Guangdong Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou 510080, Guangdong, China
| | - Yingxiong Huang
- Department of Emergency, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, China
| | - Shenming Wang
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, China.,National-Local Joint Engineering Laboratory of Vascular Disease Treatment, Guangzhou 510080, Guangdong, China.,Guangdong Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou 510080, Guangdong, China
| | - Jinsong Wang
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, China.,National-Local Joint Engineering Laboratory of Vascular Disease Treatment, Guangzhou 510080, Guangdong, China.,Guangdong Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou 510080, Guangdong, China
| |
Collapse
|
14
|
ADAM and ADAMTS disintegrin and metalloproteinases as major factors and molecular targets in vascular malfunction and disease. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 94:255-363. [PMID: 35659374 PMCID: PMC9231755 DOI: 10.1016/bs.apha.2021.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
A Disintegrin and Metalloproteinase (ADAM) and A Disintegrin and Metalloproteinase with Thrombospondin Motifs (ADAMTS) are two closely related families of proteolytic enzymes. ADAMs are largely membrane-bound enzymes that act as molecular scissors or sheddases of membrane-bound proteins, growth factors, cytokines, receptors and ligands, whereas ADAMTS are mainly secreted enzymes. ADAMs have a pro-domain, and a metalloproteinase, disintegrin, cysteine-rich and transmembrane domain. Similarly, ADAMTS family members have a pro-domain, and a metalloproteinase, disintegrin, and cysteine-rich domain, but instead of a transmembrane domain they have thrombospondin motifs. Most ADAMs and ADAMTS are activated by pro-protein convertases, and can be regulated by G-protein coupled receptor agonists, Ca2+ ionophores and protein kinase C. Activated ADAMs and ADAMTS participate in numerous vascular processes including angiogenesis, vascular smooth muscle cell proliferation and migration, vascular cell apoptosis, cell survival, tissue repair, and wound healing. ADAMs and ADAMTS also play a role in vascular malfunction and cardiovascular diseases such as hypertension, atherosclerosis, coronary artery disease, myocardial infarction, heart failure, peripheral artery disease, and vascular aneurysm. Decreased ADAMTS13 is involved in thrombotic thrombocytopenic purpura and microangiopathies. The activity of ADAMs and ADAMTS can be regulated by endogenous tissue inhibitors of metalloproteinases and other synthetic small molecule inhibitors. ADAMs and ADAMTS can be used as diagnostic biomarkers and molecular targets in cardiovascular disease, and modulators of ADAMs and ADAMTS activity may provide potential new approaches for the management of cardiovascular disorders.
Collapse
|
15
|
Wang Z, Li X, Yang J, Gong Y, Zhang H, Qiu X, Liu Y, Zhou C, Chen Y, Greenbaum J, Cheng L, Hu Y, Xie J, Yang X, Li Y, Schiller MR, Chen Y, Tan L, Tang SY, Shen H, Xiao HM, Deng HW. Single-cell RNA sequencing deconvolutes the in vivo heterogeneity of human bone marrow-derived mesenchymal stem cells. Int J Biol Sci 2021; 17:4192-4206. [PMID: 34803492 PMCID: PMC8579438 DOI: 10.7150/ijbs.61950] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 09/20/2021] [Indexed: 02/07/2023] Open
Abstract
Bone marrow-derived mesenchymal stem cells (BM-MSCs) are multipotent stromal cells that have a critical role in the maintenance of skeletal tissues such as bone, cartilage, and the fat in bone marrow. In addition to providing microenvironmental support for hematopoietic processes, BM-MSCs can differentiate into various mesodermal lineages including osteoblast/osteocyte, chondrocyte, and adipocyte that are crucial for bone metabolism. While BM-MSCs have high cell-to-cell heterogeneity in gene expression, the cell subtypes that contribute to this heterogeneity in vivo in humans have not been characterized. To investigate the transcriptional diversity of BM-MSCs, we applied single-cell RNA sequencing (scRNA-seq) on freshly isolated CD271+ BM-derived mononuclear cells (BM-MNCs) from two human subjects. We successfully identified LEPRhiCD45low BM-MSCs within the CD271+ BM-MNC population, and further codified the BM-MSCs into distinct subpopulations corresponding to the osteogenic, chondrogenic, and adipogenic differentiation trajectories, as well as terminal-stage quiescent cells. Biological functional annotations of the transcriptomes suggest that osteoblast precursors induce angiogenesis coupled with osteogenesis, and chondrocyte precursors have the potential to differentiate into myocytes. We also discovered transcripts for several clusters of differentiation (CD) markers that were either highly expressed (e.g., CD167b, CD91, CD130 and CD118) or absent (e.g., CD74, CD217, CD148 and CD68) in BM-MSCs, representing potential novel markers for human BM-MSC purification. This study is the first systematic in vivo dissection of human BM-MSCs cell subtypes at the single-cell resolution, revealing an insight into the extent of their cellular heterogeneity and roles in maintaining bone homeostasis.
Collapse
Affiliation(s)
- Zun Wang
- Xiangya School of Nursing, Central South University, Changsha, 410013, China; Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Human Normal University, Changsha, 410081, China
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, 70112, USA
| | - Xiaohua Li
- Xiangya School of Nursing, Central South University, Changsha, 410013, China; Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Human Normal University, Changsha, 410081, China
| | - Junxiao Yang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yun Gong
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, 70112, USA
| | - Huixi Zhang
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Human Normal University, Changsha, 410081, China
| | - Xiang Qiu
- School of Basic Medical Science, Central South University, Changsha, 410008, China
| | - Ying Liu
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Human Normal University, Changsha, 410081, China
| | - Cui Zhou
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Human Normal University, Changsha, 410081, China
| | - Yu Chen
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Human Normal University, Changsha, 410081, China
| | - Jonathan Greenbaum
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, 70112, USA
| | - Liang Cheng
- Department of Orthopedics and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yihe Hu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jie Xie
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Xucheng Yang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Martin R. Schiller
- Nevada Institute of Personalized Medicine and School of Life Science, 4505 S. Maryland Pkwy, Las Vegas, NV 89154-4004, USA
| | - Yiping Chen
- Department of Cell and Molecular Biology, School of Science and Engineering, Tulane University, New Orleans, LA 70112, USA
| | - Lijun Tan
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Human Normal University, Changsha, 410081, China
| | - Si-Yuan Tang
- Xiangya School of Nursing, Central South University, Changsha, 410013, China; Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Human Normal University, Changsha, 410081, China
- Hunan Women's Research Association, Changsha, 410011, China
| | - Hui Shen
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, 70112, USA
| | - Hong-Mei Xiao
- School of Basic Medical Science, Central South University, Changsha, 410008, China
- Center of Reproductive Health, System Biology and Data Information, Institute of Reproductive & Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, 410008, China
| | - Hong-Wen Deng
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, 70112, USA
- School of Basic Medical Science, Central South University, Changsha, 410008, China
| |
Collapse
|
16
|
Dyer LA, Rugonyi S. Fetal Blood Flow and Genetic Mutations in Conotruncal Congenital Heart Disease. J Cardiovasc Dev Dis 2021; 8:90. [PMID: 34436232 PMCID: PMC8397097 DOI: 10.3390/jcdd8080090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 12/19/2022] Open
Abstract
In congenital heart disease, the presence of structural defects affects blood flow in the heart and circulation. However, because the fetal circulation bypasses the lungs, fetuses with cyanotic heart defects can survive in utero but need prompt intervention to survive after birth. Tetralogy of Fallot and persistent truncus arteriosus are two of the most significant conotruncal heart defects. In both defects, blood access to the lungs is restricted or non-existent, and babies with these critical conditions need intervention right after birth. While there are known genetic mutations that lead to these critical heart defects, early perturbations in blood flow can independently lead to critical heart defects. In this paper, we start by comparing the fetal circulation with the neonatal and adult circulation, and reviewing how altered fetal blood flow can be used as a diagnostic tool to plan interventions. We then look at known factors that lead to tetralogy of Fallot and persistent truncus arteriosus: namely early perturbations in blood flow and mutations within VEGF-related pathways. The interplay between physical and genetic factors means that any one alteration can cause significant disruptions during development and underscore our need to better understand the effects of both blood flow and flow-responsive genes.
Collapse
Affiliation(s)
- Laura A. Dyer
- Department of Biology, University of Portland, Portland, OR 97203, USA;
| | - Sandra Rugonyi
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
17
|
Benedicto A, García-Kamiruaga I, Arteta B. Neuropilin-1: A feasible link between liver pathologies and COVID-19. World J Gastroenterol 2021; 27:3516-3529. [PMID: 34239266 PMCID: PMC8240058 DOI: 10.3748/wjg.v27.i24.3516] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/16/2021] [Accepted: 05/25/2021] [Indexed: 02/06/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has a tremendous impact on the health of millions of people worldwide. Unfortunately, those suffering from previous pathological conditions are more vulnerable and tend to develop more severe disease upon infection with the new SARS-CoV-2. This coronavirus interacts with the angiotensin-converting enzyme 2 receptor to invade the cells. Recently, another receptor, neuropilin-1 (NRP-1), has been reported to amplify the viral infection. Interestingly, NRP-1 is expressed in nonparenchymal liver cells and is related to and upregulated in a wide variety of liver-related pathologies. It has been observed that SARS-CoV-2 infection promotes liver injury through several pathways that may be influenced by the previous pathological status of the patient and liver expression of NRP-1. Moreover, coronavirus disease 2019 causes an inflammatory cascade called cytokine storm in patients with severe disease. This cytokine storm may influence liver sinusoidal-cell phenotype, facilitating viral invasion. In this review, the shreds of evidence linking NRP-1 with liver pathologies such as hepatocellular carcinoma, liver fibrosis, nonalcoholic fatty liver disease and inflammatory disorders are discussed in the context of SARS-CoV-2 infection. In addition, the involvement of the infection-related cytokine storm in NRP-1 overexpression and the subsequent increased risk of SARS-CoV-2 infection are also analyzed. This review aims to shed some light on the involvement of liver NRP-1 during SARS-CoV-2 infection and emphasizes the possible involvement this receptor with the observed liver damage.
Collapse
Affiliation(s)
- Aitor Benedicto
- Department of Cellular Biology and Histology, School of Medicine and Nursing, University of the Basque Country, Leioa 48940, Bizkaia, Spain
| | - Iñigo García-Kamiruaga
- Department of Gastroenterology and Hepatology, San Eloy Hospital, Barakaldo 48902, Spain
| | - Beatriz Arteta
- Department of Cellular Biology and Histology, School of Medicine and Nursing, University of the Basque Country, Leioa 48940, Bizkaia, Spain
| |
Collapse
|
18
|
Kawai T, Elliott KJ, Scalia R, Eguchi S. Contribution of ADAM17 and related ADAMs in cardiovascular diseases. Cell Mol Life Sci 2021; 78:4161-4187. [PMID: 33575814 PMCID: PMC9301870 DOI: 10.1007/s00018-021-03779-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/23/2020] [Accepted: 01/27/2021] [Indexed: 02/06/2023]
Abstract
A disintegrin and metalloproteases (ADAMs) are key mediators of cell signaling by ectodomain shedding of various growth factors, cytokines, receptors and adhesion molecules at the cellular membrane. ADAMs regulate cell proliferation, cell growth, inflammation, and other regular cellular processes. ADAM17, the most extensively studied ADAM family member, is also known as tumor necrosis factor (TNF)-α converting enzyme (TACE). ADAMs-mediated shedding of cytokines such as TNF-α orchestrates immune system or inflammatory cascades and ADAMs-mediated shedding of growth factors causes cell growth or proliferation by transactivation of the growth factor receptors including epidermal growth factor receptor. Therefore, increased ADAMs-mediated shedding can induce inflammation, tissue remodeling and dysfunction associated with various cardiovascular diseases such as hypertension and atherosclerosis, and ADAMs can be a potential therapeutic target in these diseases. In this review, we focus on the role of ADAMs in cardiovascular pathophysiology and cardiovascular diseases. The main aim of this review is to stimulate new interest in this area by highlighting remarkable evidence.
Collapse
Affiliation(s)
- Tatsuo Kawai
- Cardiovascular Research Center, Lewis Katz School of Medicine At Temple University, Philadelphia, PA, USA
| | - Katherine J Elliott
- Cardiovascular Research Center, Lewis Katz School of Medicine At Temple University, Philadelphia, PA, USA
| | - Rosario Scalia
- Cardiovascular Research Center, Lewis Katz School of Medicine At Temple University, Philadelphia, PA, USA
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine At Temple University, Philadelphia, PA, USA.
| |
Collapse
|
19
|
Chico TJA, Kugler EC. Cerebrovascular development: mechanisms and experimental approaches. Cell Mol Life Sci 2021; 78:4377-4398. [PMID: 33688979 PMCID: PMC8164590 DOI: 10.1007/s00018-021-03790-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 02/04/2021] [Accepted: 02/12/2021] [Indexed: 12/13/2022]
Abstract
The cerebral vasculature plays a central role in human health and disease and possesses several unique anatomic, functional and molecular characteristics. Despite their importance, the mechanisms that determine cerebrovascular development are less well studied than other vascular territories. This is in part due to limitations of existing models and techniques for visualisation and manipulation of the cerebral vasculature. In this review we summarise the experimental approaches used to study the cerebral vessels and the mechanisms that contribute to their development.
Collapse
Affiliation(s)
- Timothy J A Chico
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK.
- The Bateson Centre, Firth Court, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK.
- Insigneo Institute for in Silico Medicine, The Pam Liversidge Building, Sheffield, S1 3JD, UK.
| | - Elisabeth C Kugler
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK.
- The Bateson Centre, Firth Court, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK.
- Insigneo Institute for in Silico Medicine, The Pam Liversidge Building, Sheffield, S1 3JD, UK.
| |
Collapse
|
20
|
Sherafat A, Pfeiffer F, Reiss AM, Wood WM, Nishiyama A. Microglial neuropilin-1 promotes oligodendrocyte expansion during development and remyelination by trans-activating platelet-derived growth factor receptor. Nat Commun 2021; 12:2265. [PMID: 33859199 PMCID: PMC8050320 DOI: 10.1038/s41467-021-22532-2] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 03/08/2021] [Indexed: 02/02/2023] Open
Abstract
Nerve-glia (NG2) glia or oligodendrocyte precursor cells (OPCs) are distributed throughout the gray and white matter and generate myelinating cells. OPCs in white matter proliferate more than those in gray matter in response to platelet-derived growth factor AA (PDGF AA), despite similar levels of its alpha receptor (PDGFRα) on their surface. Here we show that the type 1 integral membrane protein neuropilin-1 (Nrp1) is expressed not on OPCs but on amoeboid and activated microglia in white but not gray matter in an age- and activity-dependent manner. Microglia-specific deletion of Nrp1 compromised developmental OPC proliferation in white matter as well as OPC expansion and subsequent myelin repair after acute demyelination. Exogenous Nrp1 increased PDGF AA-induced OPC proliferation and PDGFRα phosphorylation on dissociated OPCs, most prominently in the presence of suboptimum concentrations of PDGF AA. These findings uncover a mechanism of regulating oligodendrocyte lineage cell density that involves trans-activation of PDGFRα on OPCs via Nrp1 expressed by adjacent microglia.
Collapse
Affiliation(s)
- Amin Sherafat
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA
| | - Friederike Pfeiffer
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Alexander M Reiss
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA
| | - William M Wood
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA
| | - Akiko Nishiyama
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA.
- Institute for Systems Genomics, University of Connecticut, Storrs, CT, USA.
- The Connecticut Institute for the Brain and Cognitive Sciences, University of Connecticut, Storrs, CT, USA.
| |
Collapse
|
21
|
Guo Z, Mo Z. Regulation of endothelial cell differentiation in embryonic vascular development and its therapeutic potential in cardiovascular diseases. Life Sci 2021; 276:119406. [PMID: 33785330 DOI: 10.1016/j.lfs.2021.119406] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 03/05/2021] [Accepted: 03/14/2021] [Indexed: 12/17/2022]
Abstract
During vertebrate development, the cardiovascular system begins operating earlier than any other organ in the embryo. Endothelial cell (EC) forms the inner lining of blood vessels, and its extensive proliferation and migration are requisite for vasculogenesis and angiogenesis. Many aspects of cellular biology are involved in vasculogenesis and angiogenesis, including the tip versus stalk cell specification. Recently, epigenetics has attracted growing attention in regulating embryonic vascular development and controlling EC differentiation. Some proteins that regulate chromatin structure have been shown to be directly implicated in human cardiovascular diseases. Additionally, the roles of important EC signaling such as vascular endothelial growth factor and its receptors, angiopoietin-1 and tyrosine kinase containing immunoglobulin and epidermal growth factor homology domain-2, and transforming growth factor-β in EC differentiation during embryonic vasculature development are briefly discussed in this review. Recently, the transplantation of human induced pluripotent stem cell (iPSC)-ECs are promising approaches for the treatment of ischemic cardiovascular disease including myocardial infarction. Patient-specific iPSC-derived EC is a potential new target to study differences in gene expression or response to drugs. However, clinical application of the iPSC-ECs in regenerative medicine is often limited by the challenges of maintaining cell viability and function. Therefore, novel insights into the molecular mechanisms underlying EC differentiation might provide a better understanding of embryonic vascular development and bring out more effective EC-based therapeutic strategies for cardiovascular diseases.
Collapse
Affiliation(s)
- Zi Guo
- Department of Endocrinology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhaohui Mo
- Department of Endocrinology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
22
|
Moin ASM, Al-Qaissi A, Sathyapalan T, Atkin SL, Butler AE. Soluble Neuropilin-1 Response to Hypoglycemia in Type 2 Diabetes: Increased Risk or Protection in SARS-CoV-2 Infection? Front Endocrinol (Lausanne) 2021; 12:665134. [PMID: 34248841 PMCID: PMC8261232 DOI: 10.3389/fendo.2021.665134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 06/08/2021] [Indexed: 01/20/2023] Open
Abstract
INTRODUCTION Neuropilin-1(NRP1) is a cofactor that enhances SARS-CoV-2 coronavirus cell infectivity when co-expressed with angiotensin-converting enzyme 2(ACE2). The Renin-Angiotensin System (RAS) is activated in type 2 diabetes (T2D); therefore, the aim of this study was to determine if hypoglycaemia-induced stress in T2D would potentiate serum NRP1(sNRP1) levels, reflecting an increased risk for SARS-CoV-2 infection. METHODS A case-control study of aged-matched T2D (n = 23) and control (n = 23) subjects who underwent a hyperinsulinemic clamp over 1-hour to hypoglycemia(<40mg/dl) with subsequent timecourse of 4-hours and 24-hours. Slow Off-rate Modified Aptamer (SOMA)-scan plasma protein measurement determined RAS-related proteins: renin (REN), angiotensinogen (AGT), ACE2, soluble NRP1(sNRP1), NRP1 ligands (Vascular endothelial growth factor, VEGF and Class 3 Semaphorins, SEM3A) and NRP1 proteolytic enzyme (A Disintegrin and Metalloproteinase 9, ADAM9). RESULTS Baseline RAS overactivity was present with REN elevated and AGT decreased in T2D (p<0.05); ACE2 was unchanged. Baseline sNRP1, VEGF and ADAM9 did not differ between T2D and controls and remained unchanged in response to hypoglycaemia. However, 4-hours post-hypoglycemia, sNRP1, VEGF and ADAM9 were elevated in T2D(p<0.05). SEMA3A was not different at baseline; at hypoglycemia, SEMA3A decreased in controls only. Post-hypoglycemia, SEMA3A levels were higher in T2D versus controls. sNRP1 did not correlate with ACE2, REN or AGT. T2D subjects stratified according to ACE inhibitor (ACEi) therapies showed no difference in sNRP1 levels at either glucose normalization or hypoglycaemia. CONCLUSION Hypoglycemia potentiated both plasma sNRP1 level elevation and its ligands VEGF and SEMA3A, likely through an ADAM9-mediated mechanism that was not associated with RAS overactivity or ACEi therapy; however, whether this is protective or promotes increased risk for SARS-CoV-2 infection in T2D is unclear. CLINICAL TRIAL REGISTRATION https://clinicaltrials.gov, identifier NCT03102801.
Collapse
Affiliation(s)
- Abu Saleh Md Moin
- Diabetes Research Center (DRC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Ahmed Al-Qaissi
- Academic Endocrinology, Diabetes and Metabolism, Hull York Medical School, Hull, United Kingdom
- Department of Endocrinology, Leeds Medical School, Leeds, United Kingdom
| | - Thozhukat Sathyapalan
- Academic Endocrinology, Diabetes and Metabolism, Hull York Medical School, Hull, United Kingdom
| | - Stephen L. Atkin
- Department of Research, Royal College of Surgeons in Ireland Bahrain, Adliya, Bahrain
| | - Alexandra E. Butler
- Diabetes Research Center (DRC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
- *Correspondence: Alexandra E. Butler, ;
| |
Collapse
|
23
|
Chou CW, Huang YK, Kuo TT, Liu JP, Sher YP. An Overview of ADAM9: Structure, Activation, and Regulation in Human Diseases. Int J Mol Sci 2020; 21:ijms21207790. [PMID: 33096780 PMCID: PMC7590139 DOI: 10.3390/ijms21207790] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/17/2020] [Accepted: 10/19/2020] [Indexed: 12/16/2022] Open
Abstract
ADAM9 (A disintegrin and a metalloprotease 9) is a membrane-anchored protein that participates in a variety of physiological functions, primarily through the disintegrin domain for adhesion and the metalloprotease domain for ectodomain shedding of a wide variety of cell surface proteins. ADAM9 influences the developmental process, inflammation, and degenerative diseases. Recently, increasing evidence has shown that ADAM9 plays an important role in tumor biology. Overexpression of ADAM9 has been found in several cancer types and is correlated with tumor aggressiveness and poor prognosis. In addition, through either proteolytic or non-proteolytic pathways, ADAM9 promotes tumor progression, therapeutic resistance, and metastasis of cancers. Therefore, comprehensively understanding the mechanism of ADAM9 is crucial for the development of therapeutic anti-cancer strategies. In this review, we summarize the current understanding of ADAM9 in biological function, pathophysiological diseases, and various cancers. Recent advances in therapeutic strategies using ADAM9-related pathways are presented as well.
Collapse
Affiliation(s)
- Cheng-Wei Chou
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404, Taiwan; (C.-W.C.); (Y.-K.H.); (J.-P.L.)
- Department of Medicine, Division of Hematology/Medical Oncology, Taichung Veterans General Hospital, Taichung 407, Taiwan
| | - Yu-Kai Huang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404, Taiwan; (C.-W.C.); (Y.-K.H.); (J.-P.L.)
| | - Ting-Ting Kuo
- Center for Molecular Medicine, China Medical University Hospital, Taichung 404, Taiwan;
| | - Jing-Pei Liu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404, Taiwan; (C.-W.C.); (Y.-K.H.); (J.-P.L.)
| | - Yuh-Pyng Sher
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404, Taiwan; (C.-W.C.); (Y.-K.H.); (J.-P.L.)
- Center for Molecular Medicine, China Medical University Hospital, Taichung 404, Taiwan;
- Chinese Medicine Research Center, China Medical University, Taichung 404, Taiwan
- Correspondence: ; Tel.: +886-4-2205-2121
| |
Collapse
|
24
|
Chen Z, Morales JE, Avci N, Guerrero PA, Rao G, Seo JH, McCarty JH. The vascular endothelial cell-expressed prion protein doppel promotes angiogenesis and blood-brain barrier development. Development 2020; 147:dev.193094. [PMID: 32895288 DOI: 10.1242/dev.193094] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 08/17/2020] [Indexed: 12/14/2022]
Abstract
The central nervous system (CNS) contains a complex network of blood vessels that promote normal tissue development and physiology. Abnormal control of blood vessel morphogenesis and maturation is linked to the pathogenesis of various neurodevelopmental diseases. The CNS-specific genes that regulate blood vessel morphogenesis in development and disease remain largely unknown. Here, we have characterized functions for the gene encoding prion protein 2 (Prnd) in CNS blood vessel development and physiology. Prnd encodes the glycosylphosphatidylinositol (GPI)-linked protein doppel, which is expressed on the surface of angiogenic vascular endothelial cells, but is absent in quiescent endothelial cells of the adult CNS. During CNS vascular development, doppel interacts with receptor tyrosine kinases and activates cytoplasmic signaling pathways involved in endothelial cell survival, metabolism and migration. Analysis of mice genetically null for Prnd revealed impaired CNS blood vessel morphogenesis and associated endothelial cell sprouting defects. Prnd-/- mice also displayed defects in endothelial barrier integrity. Collectively, these data reveal novel mechanisms underlying doppel control of angiogenesis in the developing CNS, and may provide new insights about dysfunctional pathways that cause vascular-related CNS disorders.
Collapse
Affiliation(s)
- Zhihua Chen
- Department of Neurosurgery, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - John E Morales
- Department of Neurosurgery, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Naze Avci
- Department of Neurosurgery, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Paola A Guerrero
- Department of Neurosurgery, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Ganesh Rao
- Department of Neurosurgery, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Je Hoon Seo
- Department of Anatomy, Chungbuk National University College of Medicine, Chungbuk 28644, Republic of Korea
| | - Joseph H McCarty
- Department of Neurosurgery, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
25
|
Zhao J, Zou W, Hu T. Novel genes associated with folic acid-mediated metabolism in mouse: A bioinformatics study. PLoS One 2020; 15:e0238940. [PMID: 32915913 PMCID: PMC7485790 DOI: 10.1371/journal.pone.0238940] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 08/26/2020] [Indexed: 11/19/2022] Open
Abstract
Folic acid plays an essential role in the central nervous system and cancer. This study aimed to screen genes related to folic acid metabolism. Datasets (GSE80587, GSE65267 and GSE116299) correlated to folic acid were screened in the Gene Expression Omnibus. Weighed gene co-expression network analysis was performed to identify modules associated with sample traits of folic acid and organs (brain, prostate and kidney). Functional enrichment analysis was performed for the eigengenes in modules that were significantly correlated with sample traits. Accordingly, the hub genes and key nodes in the modules were identified using the protein interaction network. A total of 17,252 genes in three datasets were identified. One module, which included 97 genes that were highly correlated with sample traits (including folic acid treatment [cor = -0.57, P = 3e-04] and kidney [cor = -0.68, p = 4e-06]), was screened out. Hub genes, including tetratricopeptide repeat protein 38 (Ttc38) and miR-185, as well as those (including Sema3A, Insl3, Dll1, Msh4 and Snai1) associated with "neuropilin binding", "regulation of reproductive process" and "vitamin D metabolic process", were identified. Genes, including Ttc38, Sema3A, Insl3, Dll1, Msh4 and Snai1, were the novel factors that may be associated with the development of the kidneys and related to folic acid treatment.
Collapse
Affiliation(s)
- Jianwen Zhao
- Shenyang Medical College, Shenyang, Liaoning, China
| | - Wen Zou
- Liaoning Vocational College of Ecological Engineering, Shenyang, Liaoning, China
| | - Tingxi Hu
- Shenyang Medical College, Shenyang, Liaoning, China
- * E-mail:
| |
Collapse
|
26
|
Harman JL, Sayers J, Chapman C, Pellet-Many C. Emerging Roles for Neuropilin-2 in Cardiovascular Disease. Int J Mol Sci 2020; 21:E5154. [PMID: 32708258 PMCID: PMC7404143 DOI: 10.3390/ijms21145154] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 12/26/2022] Open
Abstract
Cardiovascular disease, the leading cause of death worldwide, is predominantly associated with atherosclerosis. Atherosclerosis is a chronic inflammatory disease characterised by the narrowing of large to medium-sized arteries due to a build-up of plaque. Atherosclerotic plaque is comprised of lipids, extracellular matrix, and several cell types, including endothelial, immune, and vascular smooth muscle cells. Such narrowing of the blood vessels can itself restrict blood flow to vital organs but most severe clinical complications, including heart attacks and strokes, occur when lesions rupture, triggering the blood to clot and obstructing blood flow further down the vascular tree. To circumvent such obstructions, percutaneous coronary intervention or bypass grafts are often required; however, re-occlusion of the treated artery frequently occurs. Neuropilins (NRPs), a multifunctional family of cell surface co-receptors, are expressed by endothelial, immune, and vascular smooth muscle cells and are regulators of numerous signalling pathways within the vasculature. Here, we review recent studies implicating NRP2 in the development of occlusive vascular diseases and discuss how NRP2 could be targeted for therapeutic intervention.
Collapse
Affiliation(s)
- Jennifer L Harman
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London NW1 0TU, UK
| | - Jacob Sayers
- University College London, Division of Medicine, Rayne Building, University Street, London WC1E 6JF, UK
| | - Chey Chapman
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London NW1 0TU, UK
| | - Caroline Pellet-Many
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London NW1 0TU, UK
| |
Collapse
|
27
|
Shao Y, Saredy J, Yang WY, Sun Y, Lu Y, Saaoud F, Drummer C, Johnson C, Xu K, Jiang X, Wang H, Yang X. Vascular Endothelial Cells and Innate Immunity. Arterioscler Thromb Vasc Biol 2020; 40:e138-e152. [PMID: 32459541 PMCID: PMC7263359 DOI: 10.1161/atvbaha.120.314330] [Citation(s) in RCA: 162] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In addition to the roles of endothelial cells (ECs) in physiological processes, ECs actively participate in both innate and adaptive immune responses. We previously reported that, in comparison to macrophages, a prototypic innate immune cell type, ECs have many innate immune functions that macrophages carry out, including cytokine secretion, phagocytic function, antigen presentation, pathogen-associated molecular patterns-, and danger-associated molecular patterns-sensing, proinflammatory, immune-enhancing, anti-inflammatory, immunosuppression, migration, heterogeneity, and plasticity. In this highlight, we introduce recent advances published in both ATVB and many other journals: (1) several significant characters classify ECs as novel immune cells not only in infections and allograft transplantation but also in metabolic diseases; (2) several new receptor systems including conditional danger-associated molecular pattern receptors, nonpattern receptors, and homeostasis associated molecular patterns receptors contribute to innate immune functions of ECs; (3) immunometabolism and innate immune memory determine the innate immune functions of ECs; (4) a great induction of the immune checkpoint receptors in ECs during inflammations suggests the immune tolerogenic functions of ECs; and (5) association of immune checkpoint inhibitors with cardiovascular adverse events and cardio-oncology indicates the potential contributions of ECs as innate immune cells.
Collapse
Affiliation(s)
- Ying Shao
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Jason Saredy
- Metabolic Disease Research, Cardiovascular Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - William Y. Yang
- Metabolic Disease Research, Cardiovascular Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Yu Sun
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Yifan Lu
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Fatma Saaoud
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Charles Drummer
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Candice Johnson
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Keman Xu
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Xiaohua Jiang
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
- Metabolic Disease Research, Cardiovascular Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Hong Wang
- Metabolic Disease Research, Cardiovascular Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Xiaofeng Yang
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
- Metabolic Disease Research, Cardiovascular Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| |
Collapse
|
28
|
Souza JSM, Lisboa ABP, Santos TM, Andrade MVS, Neves VBS, Teles-Souza J, Jesus HNR, Bezerra TG, Falcão VGO, Oliveira RC, Del-Bem LE. The evolution of ADAM gene family in eukaryotes. Genomics 2020; 112:3108-3116. [PMID: 32437852 DOI: 10.1016/j.ygeno.2020.05.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 03/17/2020] [Accepted: 05/08/2020] [Indexed: 12/12/2022]
Abstract
The ADAM (A Disintegrin And Metalloprotease) gene family encodes proteins with adhesion and proteolytic functions. ADAM proteins are associated with diseases like cancers. Twenty ADAM genes have been identified in humans. However, little is known about the evolution of the family. We analyzed the repertoire of ADAM genes in a vast number of eukaryotic genomes to clarify the main gene copy number expansions. For the first time, we provide compelling evidence that early-branching green algae (Mamiellophyceae) have ADAM genes, suggesting that they originated in the last common ancestor of eukaryotes, before the split of plants, fungi and animals. The ADAM family expanded in early metazoans, with the most significative gene expansion happening during the first steps of vertebrate evolution. We concluded that most of mammal ADAM diversity can be explained by gene duplications in early bone fish. Our data suggest that ADAM genes were lost early in green plant evolution.
Collapse
Affiliation(s)
- J S M Souza
- Biochemistry and Molecular Biology program, Institute of Health Sciences (ICS), Federal University of Bahia (UFBA), Salvador 40231-300, Brazil
| | - A B P Lisboa
- Biotechnology program, Institute of Health Sciences (ICS), Federal University of Bahia (UFBA), Salvador 40231-300, Brazil; Bioinformatics program, Institute of Biological Sciences (ICB), Federal University of Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil
| | - T M Santos
- Biochemistry and Molecular Biology program, Institute of Health Sciences (ICS), Federal University of Bahia (UFBA), Salvador 40231-300, Brazil; Bioinformatics program, Institute of Biological Sciences (ICB), Federal University of Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil
| | - M V S Andrade
- Biotechnology program, Institute of Health Sciences (ICS), Federal University of Bahia (UFBA), Salvador 40231-300, Brazil
| | - V B S Neves
- Biochemistry and Molecular Biology program, Institute of Health Sciences (ICS), Federal University of Bahia (UFBA), Salvador 40231-300, Brazil
| | - J Teles-Souza
- Biochemistry and Molecular Biology program, Institute of Health Sciences (ICS), Federal University of Bahia (UFBA), Salvador 40231-300, Brazil
| | - H N R Jesus
- Biochemistry and Molecular Biology program, Institute of Health Sciences (ICS), Federal University of Bahia (UFBA), Salvador 40231-300, Brazil
| | - T G Bezerra
- Biochemistry and Molecular Biology program, Institute of Health Sciences (ICS), Federal University of Bahia (UFBA), Salvador 40231-300, Brazil
| | - V G O Falcão
- Biotechnology program, Institute of Health Sciences (ICS), Federal University of Bahia (UFBA), Salvador 40231-300, Brazil
| | - R C Oliveira
- Biochemistry and Molecular Biology program, Institute of Health Sciences (ICS), Federal University of Bahia (UFBA), Salvador 40231-300, Brazil
| | - L E Del-Bem
- Institute of Health Sciences (ICS), Federal University of Bahia (UFBA), Salvador 40231-300, Brazil.
| |
Collapse
|
29
|
Neuropilin: Handyman and Power Broker in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1223:31-67. [PMID: 32030684 DOI: 10.1007/978-3-030-35582-1_3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Neuropilin-1 and neuropilin-2 form a small family of transmembrane receptors, which, due to the lack of a cytosolic protein kinase domain, act primarily as co-receptors for various ligands. Performing at the molecular level both the executive and organizing functions of a handyman as well as of a power broker, they are instrumental in controlling the signaling of various receptor tyrosine kinases, integrins, and other molecules involved in the regulation of physiological and pathological angiogenic processes. In this setting, the various neuropilin ligands and interaction partners on various cells of the tumor microenvironment, such as cancer cells, endothelial cells, cancer-associated fibroblasts, and immune cells, are surveyed. The suitability of various neuropilin-targeting substances and the intervention in neuropilin-mediated interactions is considered as a possible building block of tumor therapy.
Collapse
|
30
|
Wang Z, Liu T, Li G, Cao Z. The exploration of new therapeutic targets for HPV-negative head and neck squamous cell cancer through the construction of a ceRNA network and immune microenvironment analysis. J Cell Biochem 2020; 121:3426-3437. [PMID: 31898341 DOI: 10.1002/jcb.29615] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 12/11/2019] [Indexed: 01/11/2023]
Abstract
Previous studies have shown that human papillomavirus (HPV)-negative patients with head and neck squamous cell cancer (HNSCC) suffer from an unsatisfactory prognosis. Long noncoding RNAs (lncRNAs) have been verified to participate in many biological processes, including regulating gene expression as competing endogenous RNAs (ceRNAs), while few studies focused the ceRNA network regulation mechanism in patients with HPV-negative HNSCC tumor. Meanwhile, the immune microenvironment may be critical in the development and prognosis of HPV-negative tumors. Our study aimed to further investigate the pathogenesis and potential biomarkers for the diagnosis, therapy and prognosis of HPV-negative HNSCC through a ceRNA network. Comprehensively analyzing the sequencing data of lncRNAs, microRNAs (miRNAs), and messenger RNAs (mRNAs) in The Cancer Genome Atlas HNSCC dataset, we constructed a differentially expressed ceRNA network containing 131 lncRNAs, 35 miRNAs and 162 mRNAs. Then, survival analysis in the network was cited to explore the prognostic biomarkers. Eight mRNAs, nine lncRNAs, and one miRNA were identified to be associated with prognosis. Neuropilin (NRP) binding function, retinoid X receptor (RXR) binding, and the vascular endothelial growth factor (VEGF) signaling pathway were associated with the enrichment analysis, and they also related to the immune microenvironment. Combined with the analysis of the immune microenvironment differences, we obtained new targeted therapies using an RXR agonist, or a combination of the VEGF monoclonal antibody and an NRP antagonist, which may provide a promising future for HPV-negative HNSCC patients.
Collapse
Affiliation(s)
- Zuoyuan Wang
- Department of Forensic Pathology, School of Forensic Medicine China Medical University, Shenyang, Liaoning, China.,The First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China
| | - Tianyi Liu
- The Second Clinical College, China Medical University, Shenyang, Liaoning, China
| | - Guangqi Li
- The First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China
| | - Zhipeng Cao
- Department of Forensic Pathology, School of Forensic Medicine China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
31
|
Lu HS, Schmidt AM, Hegele RA, Mackman N, Rader DJ, Weber C, Daugherty A. Annual Report on Sex in Preclinical Studies: Arteriosclerosis, Thrombosis, and Vascular Biology Publications in 2018. Arterioscler Thromb Vasc Biol 2019; 40:e1-e9. [PMID: 31869272 DOI: 10.1161/atvbaha.119.313556] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Hong S Lu
- From the Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington (H.S.L., A.D.)
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Langone Medical Center, New York, NY (A.M.S.)
| | - Robert A Hegele
- Department of Medicine and Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada (R.A.H.)
| | - Nigel Mackman
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC (N.M.)
| | - Daniel J Rader
- Departments of Medicine and Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (D.J.R.)
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität (LMU) and German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Alan Daugherty
- From the Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington (H.S.L., A.D.)
| |
Collapse
|
32
|
Slack MA, Gordon SM. Protease Activity in Vascular Disease. Arterioscler Thromb Vasc Biol 2019; 39:e210-e218. [PMID: 31553665 PMCID: PMC6764587 DOI: 10.1161/atvbaha.119.312413] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 08/20/2019] [Indexed: 01/13/2023]
Affiliation(s)
- Megan A. Slack
- Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Scott M. Gordon
- Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky College of Medicine, Lexington, KY, USA
| |
Collapse
|
33
|
Niland S, Eble JA. Neuropilins in the Context of Tumor Vasculature. Int J Mol Sci 2019; 20:ijms20030639. [PMID: 30717262 PMCID: PMC6387129 DOI: 10.3390/ijms20030639] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/23/2019] [Accepted: 01/29/2019] [Indexed: 01/09/2023] Open
Abstract
Neuropilin-1 and Neuropilin-2 form a small family of plasma membrane spanning receptors originally identified by the binding of semaphorin and vascular endothelial growth factor. Having no cytosolic protein kinase domain, they function predominantly as co-receptors of other receptors for various ligands. As such, they critically modulate the signaling of various receptor tyrosine kinases, integrins, and other molecules involved in the regulation of physiological and pathological angiogenic processes. This review highlights the diverse neuropilin ligands and interacting partners on endothelial cells, which are relevant in the context of the tumor vasculature and the tumor microenvironment. In addition to tumor cells, the latter contains cancer-associated fibroblasts, immune cells, and endothelial cells. Based on the prevalent neuropilin-mediated interactions, the suitability of various neuropilin-targeted substances for influencing tumor angiogenesis as a possible building block of a tumor therapy is discussed.
Collapse
Affiliation(s)
- Stephan Niland
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany.
| | - Johannes A Eble
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany.
| |
Collapse
|