1
|
Bege M, Ghanem Kattoub R, Borbás A. The 20th Anniversary of Pegaptanib (MacugenTM), the First Approved Aptamer Medicine: History, Recent Advances and Future Prospects of Aptamers in Therapy. Pharmaceutics 2025; 17:394. [PMID: 40143057 PMCID: PMC11944999 DOI: 10.3390/pharmaceutics17030394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 03/15/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025] Open
Abstract
In addition to classic small-molecule drugs and modern protein-based biologics, an intriguing class of medicines is the therapeutic oligonucleotides. Most approved drugs in this category are antisense oligomers or those acting via RNA interference, both of which use base hybridization. Aptamers, also known as chemical antibodies form a smaller, yet equally interesting group of oligonucleotides that can recognize a wide range of molecular targets. Despite their high potential, only two aptamers have been approved to date, pegaptanib (MacugenTM) and avacincaptad pegol (IzervayTM), both for the treatment of age-related macular degeneration (AMD). Targeting vascular endothelial growth factor (VEGF), which plays an important role in the pathogenesis of many eye diseases, pegaptanib emerged as the first anti-VEGF agent and was used in various indications, further inspiring the development of other anti-VEGF therapies. In this review, we summarize the history of the first approved aptamer medicine, pegaptanib. We describe its chemistry and track its development from the earliest stages to the preclinical phase, clinical trials, and eventual regulatory approval. Additionally, we evaluate its position among other therapeutic agents and provide a comprehensive overview of pegaptanib's efficacy, safety, and cost-effectiveness, comparing these aspects with those of monoclonal antibodies with similar indications, bevacizumab and ranibizumab.
Collapse
Affiliation(s)
- Miklós Bege
- Department of Pharmaceutical Chemistry, University of Debrecen, Egyetem tér 1, 4032 Debrecen, Hungary; (M.B.); (R.G.K.)
| | - Rasha Ghanem Kattoub
- Department of Pharmaceutical Chemistry, University of Debrecen, Egyetem tér 1, 4032 Debrecen, Hungary; (M.B.); (R.G.K.)
- Doctoral School of Pharmaceutical Sciences, Faculty of Pharmacy, University of Debrecen, Nagyerdei krt. 98., 4032 Debrecen, Hungary
| | - Anikó Borbás
- Department of Pharmaceutical Chemistry, University of Debrecen, Egyetem tér 1, 4032 Debrecen, Hungary; (M.B.); (R.G.K.)
| |
Collapse
|
2
|
Walter LD, Orton JL, Ntekas I, Fong EHH, Maymi VI, Rudd BD, De Vlaminck I, Elisseeff JH, Cosgrove BD. Transcriptomic analysis of skeletal muscle regeneration across mouse lifespan identifies altered stem cell states. NATURE AGING 2024; 4:1862-1881. [PMID: 39578558 PMCID: PMC11645289 DOI: 10.1038/s43587-024-00756-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 10/18/2024] [Indexed: 11/24/2024]
Abstract
In aging, skeletal muscle regeneration declines due to alterations in both myogenic and non-myogenic cells and their interactions. This regenerative dysfunction is not understood comprehensively or with high spatiotemporal resolution. We collected an integrated atlas of 273,923 single-cell transcriptomes and high-resolution spatial transcriptomic maps from muscles of young, old and geriatric mice (~5, 20 and 26 months old) at multiple time points following myotoxin injury. We identified eight immune cell types that displayed accelerated or delayed dynamics by age. We observed muscle stem cell states and trajectories specific to old and geriatric muscles and evaluated their association with senescence by scoring experimentally derived and curated gene signatures in both single-cell and spatial transcriptomic data. This revealed an elevation of senescent-like muscle stem cell subsets within injury zones uniquely in aged muscles. This Resource provides a holistic portrait of the altered cellular states underlying muscle regenerative decline across mouse lifespan.
Collapse
Affiliation(s)
- Lauren D Walter
- Genetics, Genomics and Development Graduate Program, Cornell University, Ithaca, NY, USA
| | - Jessica L Orton
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Ioannis Ntekas
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | | | - Viviana I Maymi
- Department of Microbiology & Immunology, Cornell University, Ithaca, NY, USA
| | - Brian D Rudd
- Genetics, Genomics and Development Graduate Program, Cornell University, Ithaca, NY, USA
- Department of Microbiology & Immunology, Cornell University, Ithaca, NY, USA
| | - Iwijn De Vlaminck
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Jennifer H Elisseeff
- Translational Tissue Engineering Center, Wilmer Eye Institute, and Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Benjamin D Cosgrove
- Genetics, Genomics and Development Graduate Program, Cornell University, Ithaca, NY, USA.
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
3
|
Li R, Colombo M, Wang G, Rodriguez-Romera A, Benlabiod C, Jooss NJ, O’Sullivan J, Brierley CK, Clark SA, Pérez Sáez JM, Aragón Fernández P, Schoof EM, Porse B, Meng Y, Khan AO, Wen S, Dong P, Zhou W, Sousos N, Murphy L, Clarke M, Olijnik AA, C. Wong Z, Karali CS, Sirinukunwattana K, Ryou H, Norfo R, Cheng Q, Carrelha J, Ren Z, Thongjuea S, Rathinam VA, Krishnan A, Royston D, Rabinovich GA, Mead AJ, Psaila B. A proinflammatory stem cell niche drives myelofibrosis through a targetable galectin-1 axis. Sci Transl Med 2024; 16:eadj7552. [PMID: 39383242 PMCID: PMC7616771 DOI: 10.1126/scitranslmed.adj7552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 07/01/2024] [Accepted: 09/16/2024] [Indexed: 10/11/2024]
Abstract
Myeloproliferative neoplasms are stem cell-driven cancers associated with a large burden of morbidity and mortality. Most patients present with early-stage disease, but a substantial proportion progress to myelofibrosis or secondary leukemia, advanced cancers with a poor prognosis and high symptom burden. Currently, it remains difficult to predict progression, and therapies that reliably prevent or reverse fibrosis are lacking. A major bottleneck to the discovery of disease-modifying therapies has been an incomplete understanding of the interplay between perturbed cellular and molecular states. Several cell types have individually been implicated, but a comprehensive analysis of myelofibrotic bone marrow is lacking. We therefore mapped the cross-talk between bone marrow cell types in myelofibrotic bone marrow. We found that inflammation and fibrosis are orchestrated by a "quartet" of immune and stromal cell lineages, with basophils and mast cells creating a TNF signaling hub, communicating with megakaryocytes, mesenchymal stromal cells, and proinflammatory fibroblasts. We identified the β-galactoside-binding protein galectin-1 as a biomarker of progression to myelofibrosis and poor survival in multiple patient cohorts and as a promising therapeutic target, with reduced myeloproliferation and fibrosis in vitro and in vivo and improved survival after galectin-1 inhibition. In human bone marrow organoids, TNF increased galectin-1 expression, suggesting a feedback loop wherein the proinflammatory myeloproliferative neoplasm clone creates a self-reinforcing niche, fueling progression to advanced disease. This study provides a resource for studying hematopoietic cell-niche interactions, with relevance for cancer-associated inflammation and disorders of tissue fibrosis.
Collapse
Affiliation(s)
- Rong Li
- CAMS Oxford Institute; University of Oxford; Oxford, United Kingdom (UK)
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Michela Colombo
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
- Human Technopole; Milan, Italy
| | - Guanlin Wang
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
- MRC WIMM Centre for Computational Biology, University of Oxford; Oxford, United Kingdom
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology; Fudan University, Shanghai, China
- Qizhi Institute, Shanghai, China
| | - Antonio Rodriguez-Romera
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Camelia Benlabiod
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Natalie J. Jooss
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Jennifer O’Sullivan
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Charlotte K. Brierley
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Sally-Ann Clark
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Juan M. Pérez Sáez
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | | | - Erwin M. Schoof
- Department of Biotechnology and Biomedicine, Technical University of Denmark; Denmark
| | - Bo Porse
- The Finsen Laboratory, Copenhagen University Hospital; Copenhagen, Denmark
- Biotech Research and Innovation Centre, Faculty of Health Sciences, University of Copenhagen; Denmark
- Department of Clinical Medicine, University of Copenhagen; Copenhagen, Denmark
| | - Yiran Meng
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Abdullah O. Khan
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences; University of Birmingham; Birmingham, UK
| | - Sean Wen
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Pengwei Dong
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology; Fudan University, Shanghai, China
| | - Wenjiang Zhou
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology; Fudan University, Shanghai, China
| | - Nikolaos Sousos
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Lauren Murphy
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Matthew Clarke
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Aude-Anais Olijnik
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Zoë C. Wong
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Christina Simoglou Karali
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Korsuk Sirinukunwattana
- Oxford Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford; Oxford, UK
| | - Hosuk Ryou
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford; Oxford, UK
| | - Ruggiero Norfo
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Qian Cheng
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Joana Carrelha
- Haematopoietic Stem Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, University of Oxford; Oxford, UK
| | - Zemin Ren
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Supat Thongjuea
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Vijay A Rathinam
- Department of Immunology, University of Connecticut Health School of Medicine; Farmington, ConnecticutUSA
| | - Anandi Krishnan
- Stanford Cancer Institute, Stanford University School of Medicine; Stanford, California, USA
| | - Daniel Royston
- Biotech Research and Innovation Centre, Faculty of Health Sciences, University of Copenhagen; Denmark
- Oxford University Hospitals NHS Trust; Oxford, UK
| | - Gabriel A. Rabinovich
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Facultad de Ciencias Exactas, Físicas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Adam J Mead
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
- Oxford University Hospitals NHS Trust; Oxford, UK
| | - Bethan Psaila
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
- Oxford University Hospitals NHS Trust; Oxford, UK
- Ludwig Institute for Cancer Research, University of Oxford, Oxford, UK
| |
Collapse
|
4
|
Xing X, Li Z, Xu J, Chen AZ, Archer M, Wang Y, Xu M, Wang Z, Zhu M, Qin Q, Thottappillil N, Zhou M, James AW. Requirement of Pdgfrα+ cells for calvarial bone repair. Stem Cells Transl Med 2024; 13:791-802. [PMID: 38986535 PMCID: PMC11328938 DOI: 10.1093/stcltm/szae041] [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: 02/29/2024] [Accepted: 05/19/2024] [Indexed: 07/12/2024] Open
Abstract
Platelet-derived growth factor receptor α (PDGFRα) is often considered as a general marker of mesenchymal cells and fibroblasts, but also shows expression in a portion of osteoprogenitor cells. Within the skeleton, Pdgfrα+ mesenchymal cells have been identified in bone marrow and periosteum of long bones, where they play a crucial role in participating in fracture repair. A similar examination of Pdgfrα+ cells in calvarial bone healing has not been examined. Here, we utilize Pdgfrα-CreERTM;mT/mG reporter animals to examine the contribution of Pdgfrα+ mesenchymal cells to calvarial bone repair through histology and single-cell RNA sequencing (scRNA-Seq). Results showed that Pdgfrα+ mesenchymal cells are present in several cell clusters by scRNA-Seq, and by histology a dramatic increase in Pdgfrα+ cells populated the defect site at early timepoints to give rise to healed bone tissue overtime. Notably, diphtheria toxin-mediated ablation of Pdgfrα reporter+ cells resulted in significantly impaired calvarial bone healing. Our findings suggest that Pdgfrα-expressing cells within the calvarial niche play a critical role in the process of calvarial bone repair.
Collapse
Affiliation(s)
- Xin Xing
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Zhao Li
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Jiajia Xu
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Austin Z Chen
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Mary Archer
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Yiyun Wang
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Mingxin Xu
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Ziyi Wang
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Manyu Zhu
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Qizhi Qin
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Neelima Thottappillil
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Myles Zhou
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| |
Collapse
|
5
|
Punzon-Jimenez P, Machado-Lopez A, Perez-Moraga R, Llera-Oyola J, Grases D, Galvez-Viedma M, Sibai M, Satorres-Perez E, Lopez-Agullo S, Badenes R, Ferrer-Gomez C, Porta-Pardo E, Roson B, Simon C, Mas A. Effect of aging on the human myometrium at single-cell resolution. Nat Commun 2024; 15:945. [PMID: 38296945 PMCID: PMC10830479 DOI: 10.1038/s41467-024-45143-z] [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: 07/17/2023] [Accepted: 01/17/2024] [Indexed: 02/02/2024] Open
Abstract
Age-associated myometrial dysfunction can prompt complications during pregnancy and labor, which is one of the factors contributing to the 7.8-fold increase in maternal mortality in women over 40. Using single-cell/single-nucleus RNA sequencing and spatial transcriptomics, we have constructed a cellular atlas of the aging myometrium from 186,120 cells across twenty perimenopausal and postmenopausal women. We identify 23 myometrial cell subpopulations, including contractile and venous capillary cells as well as immune-modulated fibroblasts. Myometrial aging leads to fewer contractile capillary cells, a reduced level of ion channel expression in smooth muscle cells, and impaired gene expression in endothelial, smooth muscle, fibroblast, perivascular, and immune cells. We observe altered myometrial cell-to-cell communication as an aging hallmark, which associated with the loss of 25 signaling pathways, including those related to angiogenesis, tissue repair, contractility, immunity, and nervous system regulation. These insights may contribute to a better understanding of the complications faced by older individuals during pregnancy and labor.
Collapse
Affiliation(s)
- Paula Punzon-Jimenez
- Carlos Simon Foundation, Valencia, Spain
- Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain
- Department of Pediatrics, Obstetrics and Gynecology, University of Valencia, Valencia, Spain
| | - Alba Machado-Lopez
- Carlos Simon Foundation, Valencia, Spain
- Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain
| | - Raul Perez-Moraga
- Carlos Simon Foundation, Valencia, Spain
- R&D Department, Igenomix, Valencia, Spain
| | | | - Daniela Grases
- Josep Carreras Leukaemia Research Institute (IJC), Barcelona, Spain
| | | | - Mustafa Sibai
- Josep Carreras Leukaemia Research Institute (IJC), Barcelona, Spain
| | | | | | - Rafael Badenes
- Department of Surgery, University of Valencia, Valencia, Spain
- Hospital Clinico Universitario, Valencia, Spain
| | | | | | - Beatriz Roson
- Carlos Simon Foundation, Valencia, Spain
- Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain
| | - Carlos Simon
- Carlos Simon Foundation, Valencia, Spain.
- Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain.
- Department of Pediatrics, Obstetrics and Gynecology, University of Valencia, Valencia, Spain.
- Department of Obstetrics and Gynecology, BIDMC, Harvard University, Boston, MA, USA.
| | - Aymara Mas
- Carlos Simon Foundation, Valencia, Spain.
- Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain.
| |
Collapse
|
6
|
Ye RZ, Montastier E, Frisch F, Noll C, Allard-Chamard H, Gévry N, Tchernof A, Carpentier AC. Adipocyte hypertrophy associates with in vivo postprandial fatty acid metabolism and adipose single-cell transcriptional dynamics. iScience 2024; 27:108692. [PMID: 38226167 PMCID: PMC10788217 DOI: 10.1016/j.isci.2023.108692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/07/2023] [Accepted: 12/05/2023] [Indexed: 01/17/2024] Open
Abstract
Adipocyte hypertrophy is associated with metabolic complications independent of obesity. We aimed to determine: 1) the association between adipocyte size and postprandial fatty acid metabolism; 2) the potential mechanisms driving the obesity-independent, hypertrophy-associated dysmetabolism in vivo and at a single-cell resolution. Tracers with positron emission tomography were used to measure fatty acid metabolism in 40 men and women with normal or impaired glucose tolerance (NCT02808182), and single nuclei RNA-sequencing (snRNA-seq) to determine transcriptional dynamics of subcutaneous adipose tissue (AT) between individuals with AT hypertrophy vs. hyperplasia matched for sex, ethnicity, glucose-tolerance status, BMI, total and percent body fat, and waist circumference. Adipocyte size was associated with high postprandial total cardiac fatty acid uptake and higher visceral AT dietary fatty acid uptake, but lower lean tissue dietary fatty acid uptake. We found major shifts in cell transcriptomal dynamics with AT hypertrophy that were consistent with in vivo metabolic changes.
Collapse
Affiliation(s)
- Run Zhou Ye
- Division of Endocrinology, Department of Medicine, Centre de recherche du CHUS, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Emilie Montastier
- Division of Endocrinology, Department of Medicine, Centre de recherche du CHUS, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Frédérique Frisch
- Division of Endocrinology, Department of Medicine, Centre de recherche du CHUS, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Christophe Noll
- Division of Endocrinology, Department of Medicine, Centre de recherche du CHUS, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Hugues Allard-Chamard
- Division of Endocrinology, Department of Medicine, Centre de recherche du CHUS, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Nicolas Gévry
- Department of Biology, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada
| | - André Tchernof
- Québec Heart and Lung Research Institute, Laval University, Québec, QC G1V 4G5, Canada
| | - André C. Carpentier
- Division of Endocrinology, Department of Medicine, Centre de recherche du CHUS, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Department of Nuclear Medicine and Radiobiology, Centre de Recherche du CHUS, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| |
Collapse
|
7
|
Yang C, Zhang X, Yang X, Lian F, Sun Z, Huang Y, Shen W. Function and regulation of RGS family members in solid tumours: a comprehensive review. Cell Commun Signal 2023; 21:316. [PMID: 37924113 PMCID: PMC10623796 DOI: 10.1186/s12964-023-01334-7] [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: 05/10/2023] [Accepted: 09/25/2023] [Indexed: 11/06/2023] Open
Abstract
G protein-coupled receptors (GPCRs) play a key role in regulating the homeostasis of the internal environment and are closely associated with tumour progression as major mediators of cellular signalling. As a diverse and multifunctional group of proteins, the G protein signalling regulator (RGS) family was proven to be involved in the cellular transduction of GPCRs. Growing evidence has revealed dysregulation of RGS proteins as a common phenomenon and highlighted the key roles of these proteins in human cancers. Furthermore, their differential expression may be a potential biomarker for tumour diagnosis, treatment and prognosis. Most importantly, there are few systematic reviews on the functional/mechanistic characteristics and clinical application of RGS family members at present. In this review, we focus on the G-protein signalling regulator (RGS) family, which includes more than 20 family members. We analysed the classification, basic structure, and major functions of the RGS family members. Moreover, we summarize the expression changes of each RGS family member in various human cancers and their important roles in regulating cancer cell proliferation, stem cell maintenance, tumorigenesis and cancer metastasis. On this basis, we outline the molecular signalling pathways in which some RGS family members are involved in tumour progression. Finally, their potential application in the precise diagnosis, prognosis and treatment of different types of cancers and the main possible problems for clinical application at present are discussed. Our review provides a comprehensive understanding of the role and potential mechanisms of RGS in regulating tumour progression. Video Abstract.
Collapse
Affiliation(s)
- Chenglong Yang
- Key Laboratory of Precision Oncology in Universities of Shandong, Institute of Precision Medicine, Jining Medical University, Jining, 272067, China
| | - Xiaoyuan Zhang
- Key Laboratory of Precision Oncology in Universities of Shandong, Institute of Precision Medicine, Jining Medical University, Jining, 272067, China
| | - Xiaowen Yang
- Key Laboratory of Precision Oncology in Universities of Shandong, Institute of Precision Medicine, Jining Medical University, Jining, 272067, China
| | - Fuming Lian
- Key Laboratory of Precision Oncology in Universities of Shandong, Institute of Precision Medicine, Jining Medical University, Jining, 272067, China
| | - Zongrun Sun
- Key Laboratory of Precision Oncology in Universities of Shandong, Institute of Precision Medicine, Jining Medical University, Jining, 272067, China
| | - Yongming Huang
- Department of General Surgery, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272067, China.
| | - Wenzhi Shen
- Key Laboratory of Precision Oncology in Universities of Shandong, Institute of Precision Medicine, Jining Medical University, Jining, 272067, China.
| |
Collapse
|
8
|
Lu G, Du R, Liu Y, Zhang S, Li J, Pei J. RGS5 as a Biomarker of Pericytes, Involvement in Vascular Remodeling and Pulmonary Arterial Hypertension. Vasc Health Risk Manag 2023; 19:673-688. [PMID: 37881333 PMCID: PMC10596204 DOI: 10.2147/vhrm.s429535] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 10/12/2023] [Indexed: 10/27/2023] Open
Abstract
Introduction Pulmonary arterial hypertension (PAH) is a life-threatening disease characterized by a sustained rise in mean pulmonary artery pressure. Pulmonary vascular remodeling serves an important role in PAH. Identifying a key driver gene to regulate vascular remodeling of the pulmonary microvasculature is critical for PAH management. Methods Differentially expressed genes were identified using the Gene Expression Omnibus (GEO) GSE117261, GSE48149, GSE113439, GSE53408 and GSE16947 datasets. A co-expression network was constructed using weighted gene co-expression network analysis. Novel and key signatures of PAH were screened using four algorithms, including weighted gene co-expression network analysis, GEO2R analysis, support vector machines recursive feature elimination and robust rank aggregation rank analysis. Regulator of G-protein signaling 5 (RGS5), a pro-apoptotic/anti-proliferative protein, which regulate arterial tone and blood pressure in vascular smooth muscle cells. The expression of RGS5 was determined using reverse transcription-quantitative PCR (RT-qPCR) in PAH and normal mice. The location of RGS5 and pericytes was detected using immunofluorescence. Results Compared with that in the normal group, RGS5 expression was upregulated in the PAH group based on GEO and RT-qPCR analyses. RGS5 expression in single cells was enriched in pericytes in single-cell RNA sequencing analysis. RGS5 co-localization with pericytes was detected in the pulmonary microvasculature of PAH. Conclusion RGS5 regulates vascular remodeling of the pulmonary microvasculature and the occurrence of PAH through pericytes, which has provided novel ideas and strategies regarding the occurrence and innovative treatment of PAH.
Collapse
Affiliation(s)
- Guofang Lu
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi’an, 710032, People’s Republic of China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, 710032, People’s Republic of China
| | - Rui Du
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, Fourth Military Medical University, Xi’an, 710038, People’s Republic of China
| | - Yali Liu
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi’an, 710032, People’s Republic of China
| | - Shumiao Zhang
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi’an, 710032, People’s Republic of China
| | - Juan Li
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi’an, 710032, People’s Republic of China
| | - Jianming Pei
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi’an, 710032, People’s Republic of China
| |
Collapse
|
9
|
Giannone AA, Sellitto C, Rosati B, McKinnon D, White TW. Single-Cell RNA Sequencing Analysis of the Early Postnatal Mouse Lens Epithelium. Invest Ophthalmol Vis Sci 2023; 64:37. [PMID: 37870847 PMCID: PMC10599162 DOI: 10.1167/iovs.64.13.37] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 10/06/2023] [Indexed: 10/24/2023] Open
Abstract
Purpose The lens epithelium maintains the overall health of the organ. We used single-cell RNA sequencing (scRNA-seq) technology to assess transcriptional heterogeneity between cells in the postnatal day 2 (P2) epithelium and identify distinct epithelial cell subtypes. Analysis of these data was used to better understand lens growth, differentiation, and homeostasis on P2. Methods scRNA-seq on P2 mouse lenses was performed using the 10x Genomics Chromium Single Cell 3' Kit (v3.1) and short-read Illumina sequencing. Sequence alignment and preprocessing of data were conducted using 10x Genomics Cell Ranger software. Seurat was employed for preprocessing, quality control, dimensionality reduction, and cell clustering, and Monocle was utilized for trajectory analysis to understand the developmental progression of the lens cells. CellChat and GO analyses were used to explore cell-cell communication networks and signaling interactions. Results Lens epithelial cells (LECs) were divided into seven subclusters, classified by specific gene markers. The expression of crystallin, cell-cycle, and metabolic genes was not uniform, indicating distinct functional roles of LECs. Trajectory analysis predicted a bifurcation of differentiating and cycling cells from an Igfbp5+ progenitor pool. We also identified heterogeneity in signaling molecules and pathways, suggesting that cycling and progenitor subclusters have prominent roles in coordinating crosstalk. Conclusions scRNA-seq corroborated many known markers of epithelial differentiation and proliferation while providing further insight into the pathways and genes directing these processes. Interestingly, we demonstrated that the developing epithelium can be divided into distinct subpopulations. These clusters reflect the transcriptionally diverse roles of the epithelium in proliferation, signaling, and maintenance.
Collapse
Affiliation(s)
- Adrienne A. Giannone
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook University, Stony Brook, New York, United States
| | - Caterina Sellitto
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook University, Stony Brook, New York, United States
| | - Barbara Rosati
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook University, Stony Brook, New York, United States
- Veterans Affairs Medical Center, Northport, New York, United States
| | - David McKinnon
- Department of Neurobiology and Behavior, Stony Brook University School of Medicine, Stony Brook University, Stony Brook, New York, United States
| | - Thomas W. White
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook University, Stony Brook, New York, United States
| |
Collapse
|
10
|
Mateus Gonçalves L, Fahd Qadir MM, Boulina M, Makhmutova M, Pereira E, Almaça J. Pericyte dysfunction and impaired vasomotion are hallmarks of islets during the pathogenesis of type 1 diabetes. Cell Rep 2023; 42:112913. [PMID: 37531253 PMCID: PMC10529889 DOI: 10.1016/j.celrep.2023.112913] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/26/2023] [Accepted: 07/16/2023] [Indexed: 08/04/2023] Open
Abstract
Pancreatic islets are endocrine organs that depend on their microvasculature to function. Along with endothelial cells, pericytes comprise the islet microvascular network. These mural cells are crucial for microvascular stability and function, but it is not known if/how they are affected during the development of type 1 diabetes (T1D). Here, we investigate islet pericyte density, phenotype, and function using living pancreas slices from donors without diabetes, donors with a single T1D-associated autoantibody (GADA+), and recent onset T1D cases. Our data show that islet pericyte and capillary responses to vasoactive stimuli are impaired early on in T1D. Microvascular dysfunction is associated with a switch in the phenotype of islet pericytes toward myofibroblasts. Using publicly available RNA sequencing (RNA-seq) data, we further found that transcriptional alterations related to endothelin-1 signaling and vascular and extracellular matrix (ECM) remodeling are hallmarks of single autoantibody (Aab)+ donor pancreata. Our data show that microvascular dysfunction is present at early stages of islet autoimmunity.
Collapse
Affiliation(s)
- Luciana Mateus Gonçalves
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Mirza Muhammad Fahd Qadir
- Section of Endocrinology and Metabolism, John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA; Southeast Louisiana Veterans Health Care System, New Orleans, LA, USA; Tulane Center of Excellence in Sex-Based Biology & Medicine, New Orleans, LA, USA
| | - Maria Boulina
- Diabetes Research Institute, University of Miami, Miami, FL, USA
| | - Madina Makhmutova
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Elizabeth Pereira
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Joana Almaça
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA; Molecular and Cellular Pharmacology Graduate Program, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
11
|
Cherry C, Andorko JI, Krishnan K, Mejías JC, Nguyen HH, Stivers KB, Gray-Gaillard EF, Ruta A, Han J, Hamada N, Hamada M, Sturmlechner I, Trewartha S, Michel JH, Davenport Huyer L, Wolf MT, Tam AJ, Peña AN, Keerthivasan S, Le Saux CJ, Fertig EJ, Baker DJ, Housseau F, van Deursen JM, Pardoll DM, Elisseeff JH. Transfer learning in a biomaterial fibrosis model identifies in vivo senescence heterogeneity and contributions to vascularization and matrix production across species and diverse pathologies. GeroScience 2023; 45:2559-2587. [PMID: 37079217 PMCID: PMC10651581 DOI: 10.1007/s11357-023-00785-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/26/2023] [Indexed: 04/21/2023] Open
Abstract
Cellular senescence is a state of permanent growth arrest that plays an important role in wound healing, tissue fibrosis, and tumor suppression. Despite senescent cells' (SnCs) pathological role and therapeutic interest, their phenotype in vivo remains poorly defined. Here, we developed an in vivo-derived senescence signature (SenSig) using a foreign body response-driven fibrosis model in a p16-CreERT2;Ai14 reporter mouse. We identified pericytes and "cartilage-like" fibroblasts as senescent and defined cell type-specific senescence-associated secretory phenotypes (SASPs). Transfer learning and senescence scoring identified these two SnC populations along with endothelial and epithelial SnCs in new and publicly available murine and human data single-cell RNA sequencing (scRNAseq) datasets from diverse pathologies. Signaling analysis uncovered crosstalk between SnCs and myeloid cells via an IL34-CSF1R-TGFβR signaling axis, contributing to tissue balance of vascularization and matrix production. Overall, our study provides a senescence signature and a computational approach that may be broadly applied to identify SnC transcriptional profiles and SASP factors in wound healing, aging, and other pathologies.
Collapse
Affiliation(s)
- Christopher Cherry
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - James I Andorko
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kavita Krishnan
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Joscelyn C Mejías
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Helen Hieu Nguyen
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Katlin B Stivers
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elise F Gray-Gaillard
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anna Ruta
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jin Han
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Naomi Hamada
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| | - Masakazu Hamada
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| | - Ines Sturmlechner
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Pediatrics, Molecular Genetics Section, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, Netherlands
| | - Shawn Trewartha
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| | - John H Michel
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Locke Davenport Huyer
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Matthew T Wolf
- Laboratory of Cancer Immunometabolism, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Ada J Tam
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Bloomberg~Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alexis N Peña
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shilpa Keerthivasan
- Tumor Microenvironment Thematic Research Center, Bristol Myers Squibb, San Francisco, CA, USA
| | - Claude Jordan Le Saux
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Elana J Fertig
- Department of Biomedical Engineering and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD, USA
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Darren J Baker
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
- Paul F. Glenn Center for the Biology of Aging Research at Mayo Clinic, Rochester, MN, USA
| | - Franck Housseau
- Bloomberg~Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jan M van Deursen
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Drew M Pardoll
- Bloomberg~Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jennifer H Elisseeff
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Bloomberg~Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
12
|
Walter LD, Orton JL, Hannah Fong EH, Maymi VI, Rudd BD, Elisseeff JH, Cosgrove BD. Single-cell transcriptomic analysis of skeletal muscle regeneration across mouse lifespan identifies altered stem cell states associated with senescence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.25.542370. [PMID: 37292698 PMCID: PMC10245980 DOI: 10.1101/2023.05.25.542370] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Skeletal muscle regeneration is driven by the interaction of myogenic and non-myogenic cells. In aging, regeneration is impaired due to dysfunctions of myogenic and non-myogenic cells, but this is not understood comprehensively. We collected an integrated atlas of 273,923 single-cell transcriptomes from muscles of young, old, and geriatric mice (~5, 20, 26 months-old) at six time-points following myotoxin injury. We identified eight cell types, including T and NK cells and macrophage subtypes, that displayed accelerated or delayed response dynamics between ages. Through pseudotime analysis, we observed myogenic cell states and trajectories specific to old and geriatric ages. To explain these age differences, we assessed cellular senescence by scoring experimentally derived and curated gene-lists. This pointed to an elevation of senescent-like subsets specifically within the self-renewing muscle stem cells in aged muscles. This resource provides a holistic portrait of the altered cellular states underlying skeletal muscle regenerative decline across mouse lifespan.
Collapse
Affiliation(s)
- Lauren D. Walter
- Genetics, Genomics and Development Graduate Program, Cornell University, Ithaca, NY, USA
| | - Jessica L. Orton
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | | | - Viviana I. Maymi
- Department of Microbiology & Immunology, Cornell University, Ithaca, NY, USA
| | - Brian D. Rudd
- Genetics, Genomics and Development Graduate Program, Cornell University, Ithaca, NY, USA
- Department of Microbiology & Immunology, Cornell University, Ithaca, NY, USA
| | - Jennifer H. Elisseeff
- Translational Tissue Engineering Center, Wilmer Eye Institute, and Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Benjamin D. Cosgrove
- Genetics, Genomics and Development Graduate Program, Cornell University, Ithaca, NY, USA
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| |
Collapse
|
13
|
Delcroix V, Mauduit O, Lee HS, Ivanova A, Umazume T, Knox SM, de Paiva CS, Dartt DA, Makarenkova HP. The First Transcriptomic Atlas of the Adult Lacrimal Gland Reveals Epithelial Complexity and Identifies Novel Progenitor Cells in Mice. Cells 2023; 12:1435. [PMID: 37408269 PMCID: PMC10216974 DOI: 10.3390/cells12101435] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/13/2023] [Accepted: 05/16/2023] [Indexed: 07/07/2023] Open
Abstract
The lacrimal gland (LG) secretes aqueous tears. Previous studies have provided insights into the cell lineage relationships during tissue morphogenesis. However, little is known about the cell types composing the adult LG and their progenitors. Using scRNAseq, we established the first comprehensive cell atlas of the adult mouse LG to investigate the cell hierarchy, its secretory repertoire, and the sex differences. Our analysis uncovered the complexity of the stromal landscape. Epithelium subclustering revealed myoepithelial cells, acinar subsets, and two novel acinar subpopulations: Tfrchi and Car6hi cells. The ductal compartment contained Wfdc2+ multilayered ducts and an Ltf+ cluster formed by luminal and intercalated duct cells. Kit+ progenitors were identified as: Krt14+ basal ductal cells, Aldh1a1+ cells of Ltf+ ducts, and Sox10+ cells of the Car6hi acinar and Ltf+ epithelial clusters. Lineage tracing experiments revealed that the Sox10+ adult populations contribute to the myoepithelial, acinar, and ductal lineages. Using scRNAseq data, we found that the postnatally developing LG epithelium harbored key features of putative adult progenitors. Finally, we showed that acinar cells produce most of the sex-biased lipocalins and secretoglobins detected in mouse tears. Our study provides a wealth of new data on LG maintenance and identifies the cellular origin of sex-biased tear components.
Collapse
Affiliation(s)
- Vanessa Delcroix
- Department of Molecular and Experimental Medicine, Scripps Research Institute, La Jolla, CA 92037, USA; (V.D.); (H.S.L.); (A.I.); (T.U.)
| | - Olivier Mauduit
- Department of Molecular and Experimental Medicine, Scripps Research Institute, La Jolla, CA 92037, USA; (V.D.); (H.S.L.); (A.I.); (T.U.)
| | - Hyun Soo Lee
- Department of Molecular and Experimental Medicine, Scripps Research Institute, La Jolla, CA 92037, USA; (V.D.); (H.S.L.); (A.I.); (T.U.)
- Department of Ophthalmology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Anastasiia Ivanova
- Department of Molecular and Experimental Medicine, Scripps Research Institute, La Jolla, CA 92037, USA; (V.D.); (H.S.L.); (A.I.); (T.U.)
| | - Takeshi Umazume
- Department of Molecular and Experimental Medicine, Scripps Research Institute, La Jolla, CA 92037, USA; (V.D.); (H.S.L.); (A.I.); (T.U.)
| | - Sarah M. Knox
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA 94143, USA;
- Program in Craniofacial Biology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Cintia S. de Paiva
- The Ocular Surface Center, Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Darlene A. Dartt
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA;
| | - Helen P. Makarenkova
- Department of Molecular and Experimental Medicine, Scripps Research Institute, La Jolla, CA 92037, USA; (V.D.); (H.S.L.); (A.I.); (T.U.)
| |
Collapse
|
14
|
Root SH, Vrhovac Madunic I, Kronenberg MS, Cao Y, Novak S, Kalajzic I. Lineage Tracing of RGS5-CreER-Labeled Cells in Long Bones During Homeostasis and Injury. Stem Cells 2023; 41:493-504. [PMID: 36888549 PMCID: PMC10183968 DOI: 10.1093/stmcls/sxad020] [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: 10/04/2022] [Accepted: 02/21/2023] [Indexed: 03/09/2023]
Abstract
Regulator of G protein signaling 5 (RGS5) is a GTPase activator for heterotrimeric G-protein α-subunits, shown to be a marker of pericytes. Bone marrow stromal cell population (BMSCs) is heterogeneous. Populations of mesenchymal progenitors, cells supportive of hematopoiesis, and stromal cells regulating bone remodeling have been recently identified. Periosteal and bone marrow mesenchymal stem cells (MSCs) are participating in fracture healing, but it is difficult to distinguish the source of cells within the callus. Considering that perivascular cells exert osteoprogenitor potential, we generated an RGS5 transgenic mouse model (Rgs5-CreER) which when crossed with Ai9 reporter animals (Rgs5/Tomato), is suitable for lineage tracing during growth and post-injury. Flow cytometry analysis and histology confirmed the presence of Rgs5/Tomato+ cells within CD31+ endothelial, CD45+ hematopoietic, and CD31-CD45- mesenchymal/perivascular cells. A tamoxifen chase showed expansion of Rgs5/Tomato+ cells expressing osterix within the trabeculae positioned between mineralized matrix and vasculature. Long-term chase showed proportion of Rgs5/Tomato+ cells contributes to mature osteoblasts expressing osteocalcin. Following femoral fracture, Rgs5/Tomato+ cells are observed around newly formed bone within the BM cavity and expressed osterix and osteocalcin, while contribution within periosteum was low and limited to fibroblastic callus with very few positive chondrocytes. In addition, BM injury model confirmed that RGS5-Cre labels population of BMSCs expands during injury and participates in osteogenesis. Under homeostatic conditions, lineage-traced RGS5 cells within the trabecular area demonstrate osteoprogenitor capacity that in an injury model contributes to new bone formation primarily within the BM niche.
Collapse
Affiliation(s)
- Sierra H Root
- Center for Regenerative Medicine and Skeletal Development, UConn Health, Farmington, CT, USA
| | - Ivana Vrhovac Madunic
- Center for Regenerative Medicine and Skeletal Development, UConn Health, Farmington, CT, USA
| | - Mark S Kronenberg
- Center for Regenerative Medicine and Skeletal Development, UConn Health, Farmington, CT, USA
| | - Ye Cao
- Center for Regenerative Medicine and Skeletal Development, UConn Health, Farmington, CT, USA
| | - Sanja Novak
- Center for Regenerative Medicine and Skeletal Development, UConn Health, Farmington, CT, USA
| | - Ivo Kalajzic
- Center for Regenerative Medicine and Skeletal Development, UConn Health, Farmington, CT, USA
| |
Collapse
|
15
|
Monavarfeshani A, Yan W, Pappas C, Odenigbo KA, He Z, Segrè AV, van Zyl T, Hageman GS, Sanes JR. Transcriptomic Analysis of the Ocular Posterior Segment Completes a Cell Atlas of the Human Eye. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.26.538447. [PMID: 37162855 PMCID: PMC10168356 DOI: 10.1101/2023.04.26.538447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Although the visual system extends through the brain, most vision loss originates from defects in the eye. Its central element is the neural retina, which senses light, processes visual signals, and transmits them to the rest of the brain through the optic nerve (ON). Surrounding the retina are numerous other structures, conventionally divided into anterior and posterior segments. Here we used high-throughput single nucleus RNA sequencing (snRNA-seq) to classify and characterize cells in the extraretinal components of the posterior segment: ON, optic nerve head (ONH), peripheral sclera, peripapillary sclera (PPS), choroid, and retinal pigment epithelium (RPE). Defects in each of these tissues are associated with blinding diseases - for example, glaucoma (ONH and PPS), optic neuritis (ON), retinitis pigmentosa (RPE), and age-related macular degeneration (RPE and choroid). From ∼151,000 single nuclei, we identified 37 transcriptomically distinct cell types, including multiple types of astrocytes, oligodendrocytes, fibroblasts, and vascular endothelial cells. Our analyses revealed a differential distribution of many cell types among distinct structures. Together with our previous analyses of the anterior segment and retina, the new data complete a "Version 1" cell atlas of the human eye. We used this atlas to map the expression of >180 genes associated with the risk of developing glaucoma, which is known to involve ocular tissues in both anterior and posterior segments as well as neural retina. Similar methods can be used to investigate numerous additional ocular diseases, many of which are currently untreatable.
Collapse
Affiliation(s)
- Aboozar Monavarfeshani
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138
- F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA, USA
- Equal contributions
| | - Wenjun Yan
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138
- Equal contributions
| | - Christian Pappas
- Sharon Eccles Steele Center for Translational Medicine, John A. Moran Eye Center, Department of Ophthalmology & Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT, 84132, USA
| | - Kenechukwu A. Odenigbo
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138
| | - Zhigang He
- F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA, USA
| | - Ayellet V. Segrè
- Department of Ophthalmology, Harvard Medical School and Massachusetts Eye and Ear, Boston, MA 02114
- Broad Institute of Harvard and MIT, Cambridge, MA
| | - Tavé van Zyl
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138
- Department of Ophthalmology, Harvard Medical School and Massachusetts Eye and Ear, Boston, MA 02114
- Present address: Department of Ophthalmology and Visual Science, Yale School of Medicine, New Haven, CT 06510
| | - Gregory S. Hageman
- Sharon Eccles Steele Center for Translational Medicine, John A. Moran Eye Center, Department of Ophthalmology & Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT, 84132, USA
| | - Joshua R. Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138
| |
Collapse
|
16
|
Mughal A, Nelson MT, Hill-Eubanks D. The post-arteriole transitional zone: a specialized capillary region that regulates blood flow within the CNS microvasculature. J Physiol 2023; 601:889-901. [PMID: 36751860 PMCID: PMC9992301 DOI: 10.1113/jp282246] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 01/13/2023] [Indexed: 02/09/2023] Open
Abstract
The brain is an energy hog, consuming available energy supplies at a rate out of all proportion to its relatively small size. This outsized demand, largely reflecting the unique computational activity of the brain, is met by an ensemble of neurovascular coupling mechanisms that link neuronal activity with local increases in blood delivery. This just-in-time replenishment strategy, made necessary by the limited energy-storage capacity of neurons, complicates the nutrient-delivery task of the cerebral vasculature, layering on a temporo-spatial requirement that invites - and challenges - mechanistic interpretation. The centre of gravity of research efforts to disentangle these mechanisms has shifted from an initial emphasis on astrocyte-arteriole-level processes to mechanisms that operate on the capillary level, a shift that has brought into sharp focus questions regarding the fine control of blood distribution to active neurons. As these investigations have drilled down into finer reaches of the microvasculature, they have revealed an arteriole-proximate subregion of CNS capillary networks that serves a regulatory function in directing blood flow into and within downstream capillaries. They have also illuminated differences in researchers' perspectives on the vascular structures and identity of mural cells in this region that impart the vasomodulatory effects that control blood distribution. In this review, we highlight the regulatory role of a variably named region of the microvasculature, referred to here as the post-arteriole transition zone, in channeling blood flow within CNS capillary networks, and underscore the contribution of dynamically contractile perivascular mural cell - generally, but not universally, recognized as pericytes - to this function.
Collapse
Affiliation(s)
- Amreen Mughal
- Department of Pharmacology, University of Vermont, Burlington, VT, USA
| | - Mark T. Nelson
- Department of Pharmacology, University of Vermont, Burlington, VT, USA
- Division of Cardiovascular Sciences, University of Manchester, Manchester, UK
| | | |
Collapse
|
17
|
Bischoff P, Trinks A, Wiederspahn J, Obermayer B, Pett JP, Jurmeister P, Elsner A, Dziodzio T, Rückert JC, Neudecker J, Falk C, Beule D, Sers C, Morkel M, Horst D, Klauschen F, Blüthgen N. The single-cell transcriptional landscape of lung carcinoid tumors. Int J Cancer 2022; 150:2058-2071. [PMID: 35262195 DOI: 10.1002/ijc.33995] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/17/2022] [Accepted: 02/22/2022] [Indexed: 11/11/2022]
Abstract
Lung carcinoid tumors, also referred to as pulmonary neuroendocrine tumors or lung carcinoids, are rare neoplasms of the lung with a more favorable prognosis than other subtypes of lung cancer. Still, some patients suffer from relapsed disease and metastatic spread. Several recent single-cell studies have provided detailed insights into the cellular heterogeneity of more common lung cancers, such as adeno- and squamous cell carcinoma. However, the characteristics of lung carcinoids on the single-cell level are yet completely unknown. To study the cellular composition and single-cell gene expression profiles in lung carcinoids, we applied single-cell RNA sequencing to three lung carcinoid tumor samples and normal lung tissue. The single-cell transcriptomes of carcinoid tumor cells reflected intertumoral heterogeneity associated with clinicopathological features, such as tumor necrosis and proliferation index. The immune microenvironment was specifically enriched in non-inflammatory monocyte-derived myeloid cells. Tumor-associated endothelial cells were characterized by distinct gene expression profiles. A spectrum of vascular smooth muscle cells and pericytes predominated the stromal microenvironment. We found a small proportion of myofibroblasts exhibiting features reminiscent of cancer-associated fibroblasts. Stromal and immune cells exhibited potential paracrine interactions which may shape the microenvironment via NOTCH, VEGF, TGFβ and JAK/STAT signaling. Moreover, single-cell gene signatures of pericytes and myofibroblasts demonstrated prognostic value in bulk gene expression data. Here, we provide first comprehensive insights into the cellular composition and single-cell gene expression profiles in lung carcinoids, demonstrating the non-inflammatory and vessel-rich nature of their tumor microenvironment, and outlining relevant intercellular interactions which could serve as future therapeutic targets. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Philip Bischoff
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Berlin, Germany.,Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.,German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Alexandra Trinks
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Berlin, Germany.,Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.,BIH Bioportal Single Cells, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jennifer Wiederspahn
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Berlin, Germany.,IRI Life Sciences, Humboldt University of Berlin, Berlin, Germany
| | - Benedikt Obermayer
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Core Unit Bioinformatics, Berlin, Germany
| | - Jan Patrick Pett
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Core Unit Bioinformatics, Berlin, Germany
| | - Philipp Jurmeister
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Berlin, Germany.,Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.,Institute of Pathology, LMU Munich, München, Germany
| | - Aron Elsner
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Surgery, Campus Charité Mitte and Campus Virchow-Klinikum, Berlin, Germany
| | - Tomasz Dziodzio
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Surgery, Campus Charité Mitte and Campus Virchow-Klinikum, Berlin, Germany
| | - Jens-Carsten Rückert
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Surgery, Campus Charité Mitte and Campus Virchow-Klinikum, Berlin, Germany
| | - Jens Neudecker
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Surgery, Campus Charité Mitte and Campus Virchow-Klinikum, Berlin, Germany
| | - Christine Falk
- Institute of Transplant Immunology, Hannover Medical School, Hannover, Germany.,DZIF, German Center for Infectious Diseases, TTU-IICH Hannover-Braunschweig site, Braunschweig, Germany
| | - Dieter Beule
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Core Unit Bioinformatics, Berlin, Germany
| | - Christine Sers
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Berlin, Germany.,Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.,German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Markus Morkel
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Berlin, Germany.,Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.,German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,BIH Bioportal Single Cells, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - David Horst
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Berlin, Germany.,Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.,German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Frederick Klauschen
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Berlin, Germany.,German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Institute of Pathology, LMU Munich, München, Germany
| | - Nils Blüthgen
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Berlin, Germany.,Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.,German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,IRI Life Sciences, Humboldt University of Berlin, Berlin, Germany
| |
Collapse
|
18
|
Lin X, Chi D, Meng Q, Gong Q, Tong Z. Single-Cell Sequencing Unveils the Heterogeneity of Nonimmune Cells in Chronic Apical Periodontitis. Front Cell Dev Biol 2022; 9:820274. [PMID: 35237614 PMCID: PMC8883837 DOI: 10.3389/fcell.2021.820274] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 12/24/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic apical periodontitis (CAP) is a unique dynamic interaction between microbial invasions and host defense mechanisms, resulting in infiltration of immune cells, bone absorption, and periapical granuloma formation. To help to understand periapical tissue pathophysiology, we constituted a single-cell atlas for 26,737 high-quality cells from inflammatory periapical tissue and uncovered the complex cellular landscape. The eight types of cells, including nonimmune cells and immune cells, were identified in the periapical tissue of CAP. Considering the key roles of nonimmune cells in CAP, we emphasized osteo-like cells, basal/stromal cells, endothelial cells, and epithelial cells, and discovered their diversity and heterogeneity. The temporal profiling of genomic alterations from common CAP to typical periapical granuloma provided predictions for transcription factors and biological processes. Our study presented potential clues that the shift of inflammatory cytokines, chemokines, proteases, and growth factors initiated polymorphic cell differentiation, lymphangiogenesis, and angiogenesis during CAP.
Collapse
Affiliation(s)
- Xinwei Lin
- Department of Operative Dentistry and Endodontics, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Danlu Chi
- Department of Operative Dentistry and Endodontics, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Qingzhen Meng
- Department of Operative Dentistry and Endodontics, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Qimei Gong
- Department of Operative Dentistry and Endodontics, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Qimei Gong, ; Zhongchun Tong,
| | - Zhongchun Tong
- Department of Operative Dentistry and Endodontics, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Qimei Gong, ; Zhongchun Tong,
| |
Collapse
|
19
|
Tobal R, Potjewijd J, van Empel VPM, Ysermans R, Schurgers LJ, Reutelingsperger CP, Damoiseaux JGMC, van Paassen P. Vascular Remodeling in Pulmonary Arterial Hypertension: The Potential Involvement of Innate and Adaptive Immunity. Front Med (Lausanne) 2022; 8:806899. [PMID: 35004784 PMCID: PMC8727487 DOI: 10.3389/fmed.2021.806899] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 12/02/2021] [Indexed: 11/30/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe disease with high morbidity and mortality. Current therapies are mainly focused on vasodilative agents to improve prognosis. However, recent literature has shown the important interaction between immune cells and stromal vascular cells in the pathogenic modifications of the pulmonary vasculature. The immunological pathogenesis of PAH is known as a complex interplay between immune cells and vascular stromal cells, via direct contacts and/or their production of extra-cellular/diffusible factors such as cytokines, chemokines, and growth factors. These include, the B-cell—mast-cell axis, endothelium mediated fibroblast activation and subsequent M2 macrophage polarization, anti-endothelial cell antibodies and the versatile role of IL-6 on vascular cells. This review aims to outline the major pathophysiological changes in vascular cells caused by immunological mechanisms, leading to vascular remodeling, increased pulmonary vascular resistance and eventually PAH. Considering the underlying immunological mechanisms, these mechanisms may be key to halt progression of disease.
Collapse
Affiliation(s)
- Rachid Tobal
- Division of Nephrology and Clinical and Experimental Immunology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, Netherlands
| | - Judith Potjewijd
- Division of Nephrology and Clinical and Experimental Immunology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, Netherlands
| | - Vanessa P M van Empel
- Department of Cardiology, Maastricht University Medical Center, Maastricht, Netherlands
| | - Renee Ysermans
- Division of Nephrology and Clinical and Experimental Immunology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, Netherlands
| | - Leon J Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht, Netherlands
| | - Chris P Reutelingsperger
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht, Netherlands
| | - Jan G M C Damoiseaux
- Central Diagnostic Laboratory, Maastricht University Medical Center, Maastricht, Netherlands
| | - Pieter van Paassen
- Division of Nephrology and Clinical and Experimental Immunology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, Netherlands
| |
Collapse
|
20
|
Kahn BM, Lucas A, Alur RG, Wengyn MD, Schwartz GW, Li J, Sun K, Maurer HC, Olive KP, Faryabi RB, Stanger BZ. The vascular landscape of human cancer. J Clin Invest 2021; 131:136655. [PMID: 33258803 DOI: 10.1172/jci136655] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 11/12/2020] [Indexed: 02/06/2023] Open
Abstract
Tumors depend on a blood supply to deliver oxygen and nutrients, making tumor vasculature an attractive anticancer target. However, only a fraction of patients with cancer benefit from angiogenesis inhibitors. Whether antiangiogenic therapy would be more effective if targeted to individuals with specific tumor characteristics is unknown. To better characterize the tumor vascular environment both within and between cancer types, we developed a standardized metric - the endothelial index (EI) - to estimate vascular density in over 10,000 human tumors, corresponding to 31 solid tumor types, from transcriptome data. We then used this index to compare hyper- and hypovascular tumors, enabling the classification of human tumors into 6 vascular microenvironment signatures (VMSs) based on the expression of a panel of 24 vascular "hub" genes. The EI and VMS correlated with known tumor vascular features and were independently associated with prognosis in certain cancer types. Retrospective testing of clinical trial data identified VMS2 classification as a powerful biomarker for response to bevacizumab. Thus, we believe our studies provide an unbiased picture of human tumor vasculature that may enable more precise deployment of antiangiogenesis therapy.
Collapse
Affiliation(s)
- Benjamin M Kahn
- Department of Medicine.,Department of Cell and Developmental Biology.,Abramson Family Cancer Research Institute.,Abramson Cancer Center
| | - Alfredo Lucas
- Department of Medicine.,Department of Cell and Developmental Biology.,Abramson Family Cancer Research Institute.,Abramson Cancer Center
| | - Rohan G Alur
- Department of Medicine.,Department of Cell and Developmental Biology.,Abramson Family Cancer Research Institute.,Abramson Cancer Center
| | - Maximillian D Wengyn
- Department of Medicine.,Department of Cell and Developmental Biology.,Abramson Family Cancer Research Institute.,Abramson Cancer Center
| | - Gregory W Schwartz
- Abramson Family Cancer Research Institute.,Abramson Cancer Center.,Department of Pathology and Laboratory Medicine.,Penn Epigenetics Institute, and.,Department of Cancer Biology Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jinyang Li
- Department of Medicine.,Department of Cell and Developmental Biology.,Abramson Family Cancer Research Institute.,Abramson Cancer Center
| | - Kathryn Sun
- Department of Medicine.,Department of Cell and Developmental Biology.,Abramson Family Cancer Research Institute.,Abramson Cancer Center
| | - H Carlo Maurer
- Department of Medicine, Division of Digestive Liver Diseases and Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York, USA
| | - Kenneth P Olive
- Department of Medicine, Division of Digestive Liver Diseases and Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York, USA
| | - Robert B Faryabi
- Abramson Family Cancer Research Institute.,Abramson Cancer Center.,Department of Pathology and Laboratory Medicine.,Penn Epigenetics Institute, and.,Department of Cancer Biology Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ben Z Stanger
- Department of Medicine.,Department of Cell and Developmental Biology.,Abramson Family Cancer Research Institute.,Abramson Cancer Center
| |
Collapse
|
21
|
Subretinal fibrosis in neovascular age-related macular degeneration: current concepts, therapeutic avenues, and future perspectives. Cell Tissue Res 2021; 387:361-375. [PMID: 34477966 PMCID: PMC8975778 DOI: 10.1007/s00441-021-03514-8] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 07/19/2021] [Indexed: 12/13/2022]
Abstract
Age-related macular degeneration (AMD) is a progressive, degenerative disease of the human retina which in its most aggressive form is associated with the formation of macular neovascularization (MNV) and subretinal fibrosis leading to irreversible blindness. MNVs contain blood vessels as well as infiltrating immune cells, myofibroblasts, and excessive amounts of extracellular matrix proteins such as collagens, fibronectin, and laminin which disrupts retinal function and triggers neurodegeneration. In the mammalian retina, damaged neurons cannot be replaced by tissue regeneration, and subretinal MNV and fibrosis persist and thus fuel degeneration and visual loss. This review provides an overview of subretinal fibrosis in neovascular AMD, by summarizing its clinical manifestations, exploring the current understanding of the underlying cellular and molecular mechanisms and discussing potential therapeutic approaches to inhibit subretinal fibrosis in the future.
Collapse
|
22
|
Donadon M, Santoro MM. The origin and mechanisms of smooth muscle cell development in vertebrates. Development 2021; 148:148/7/dev197384. [PMID: 33789914 DOI: 10.1242/dev.197384] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Smooth muscle cells (SMCs) represent a major structural and functional component of many organs during embryonic development and adulthood. These cells are a crucial component of vertebrate structure and physiology, and an updated overview of the developmental and functional process of smooth muscle during organogenesis is desirable. Here, we describe the developmental origin of SMCs within different tissues by comparing their specification and differentiation with other organs, including the cardiovascular, respiratory and intestinal systems. We then discuss the instructive roles of smooth muscle in the development of such organs through signaling and mechanical feedback mechanisms. By understanding SMC development, we hope to advance therapeutic approaches related to tissue regeneration and other smooth muscle-related diseases.
Collapse
Affiliation(s)
- Michael Donadon
- Department of Biology, University of Padua, Via U. Bassi 58B, 35121 Padua, Italy
| | - Massimo M Santoro
- Department of Biology, University of Padua, Via U. Bassi 58B, 35121 Padua, Italy
| |
Collapse
|
23
|
Girolamo F, de Trizio I, Errede M, Longo G, d'Amati A, Virgintino D. Neural crest cell-derived pericytes act as pro-angiogenic cells in human neocortex development and gliomas. Fluids Barriers CNS 2021; 18:14. [PMID: 33743764 PMCID: PMC7980348 DOI: 10.1186/s12987-021-00242-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 02/13/2021] [Indexed: 02/07/2023] Open
Abstract
Central nervous system diseases involving the parenchymal microvessels are frequently associated with a ‘microvasculopathy’, which includes different levels of neurovascular unit (NVU) dysfunction, including blood–brain barrier alterations. To contribute to the understanding of NVU responses to pathological noxae, we have focused on one of its cellular components, the microvascular pericytes, highlighting unique features of brain pericytes with the aid of the analyses carried out during vascularization of human developing neocortex and in human gliomas. Thanks to their position, centred within the endothelial/glial partition of the vessel basal lamina and therefore inserted between endothelial cells and the perivascular and vessel-associated components (astrocytes, oligodendrocyte precursor cells (OPCs)/NG2-glia, microglia, macrophages, nerve terminals), pericytes fulfil a central role within the microvessel NVU. Indeed, at this critical site, pericytes have a number of direct and extracellular matrix molecule- and soluble factor-mediated functions, displaying marked phenotypical and functional heterogeneity and carrying out multitasking services. This pericytes heterogeneity is primarily linked to their position in specific tissue and organ microenvironments and, most importantly, to their ontogeny. During ontogenesis, pericyte subtypes belong to two main embryonic germ layers, mesoderm and (neuro)ectoderm, and are therefore expected to be found in organs ontogenetically different, nonetheless, pericytes of different origin may converge and colonize neighbouring areas of the same organ/apparatus. Here, we provide a brief overview of the unusual roles played by forebrain pericytes in the processes of angiogenesis and barriergenesis by virtue of their origin from midbrain neural crest stem cells. A better knowledge of the ontogenetic subpopulations may support the understanding of specific interactions and mechanisms involved in pericyte function/dysfunction, including normal and pathological angiogenesis, thereby offering an alternative perspective on cell subtype-specific therapeutic approaches. ![]()
Collapse
Affiliation(s)
- Francesco Girolamo
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy.
| | - Ignazio de Trizio
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy.,Intensive Care Unit, Department of Intensive Care, Regional Hospital of Lugano, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Mariella Errede
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy
| | - Giovanna Longo
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Molecular Biology Unit, University of Bari School of Medicine, Bari, Italy
| | - Antonio d'Amati
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy.,Department of Emergency and Organ Transplantation, Pathology Section, University of Bari School of Medicine, Bari, Italy
| | - Daniela Virgintino
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy
| |
Collapse
|
24
|
Girolamo F, de Trizio I, Errede M, Longo G, d’Amati A, Virgintino D. Neural crest cell-derived pericytes act as pro-angiogenic cells in human neocortex development and gliomas. Fluids Barriers CNS 2021. [DOI: 10.1186/s12987-021-00242-7 union select null--] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
AbstractCentral nervous system diseases involving the parenchymal microvessels are frequently associated with a ‘microvasculopathy’, which includes different levels of neurovascular unit (NVU) dysfunction, including blood–brain barrier alterations. To contribute to the understanding of NVU responses to pathological noxae, we have focused on one of its cellular components, the microvascular pericytes, highlighting unique features of brain pericytes with the aid of the analyses carried out during vascularization of human developing neocortex and in human gliomas. Thanks to their position, centred within the endothelial/glial partition of the vessel basal lamina and therefore inserted between endothelial cells and the perivascular and vessel-associated components (astrocytes, oligodendrocyte precursor cells (OPCs)/NG2-glia, microglia, macrophages, nerve terminals), pericytes fulfil a central role within the microvessel NVU. Indeed, at this critical site, pericytes have a number of direct and extracellular matrix molecule- and soluble factor-mediated functions, displaying marked phenotypical and functional heterogeneity and carrying out multitasking services. This pericytes heterogeneity is primarily linked to their position in specific tissue and organ microenvironments and, most importantly, to their ontogeny. During ontogenesis, pericyte subtypes belong to two main embryonic germ layers, mesoderm and (neuro)ectoderm, and are therefore expected to be found in organs ontogenetically different, nonetheless, pericytes of different origin may converge and colonize neighbouring areas of the same organ/apparatus. Here, we provide a brief overview of the unusual roles played by forebrain pericytes in the processes of angiogenesis and barriergenesis by virtue of their origin from midbrain neural crest stem cells. A better knowledge of the ontogenetic subpopulations may support the understanding of specific interactions and mechanisms involved in pericyte function/dysfunction, including normal and pathological angiogenesis, thereby offering an alternative perspective on cell subtype-specific therapeutic approaches.
Collapse
|
25
|
Fibrotic Changes and Endothelial-to-Mesenchymal Transition Promoted by VEGFR2 Antagonism Alter the Therapeutic Effects of VEGFA Pathway Blockage in a Mouse Model of Choroidal Neovascularization. Cells 2020; 9:cells9092057. [PMID: 32917003 PMCID: PMC7563259 DOI: 10.3390/cells9092057] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/08/2020] [Accepted: 09/08/2020] [Indexed: 12/15/2022] Open
Abstract
Many patients with wet age-related macular degeneration do not respond well to anti- vascular endothelial growth factor A (VEGFA) therapy for choroidal neovascularization (CNV), and the efficacy of anti-VEGFA decreases over time. We investigated the hypothesis that fibrotic changes, in particular via endothelial-to-mesenchymal transition (EndoMT), play a role in CNV and alter the therapeutic effects of VEGFA pathway blockage. Induction of EndoMT of primary human retinal endothelial cells led to a significantly reduced response to VEGFA at the level of gene expression, cellular proliferation, migration, and tube formation. Suppression of EndoMT restored cell responsiveness to VEGFA. In a mouse model of spontaneous CNV, fibrotic changes and EndoMT persisted as the CNV lesions became more established over time. VEGFA receptor-2 (VEGFR2) antagonism further induced fibrosis and EndoMT in the CNV. The combination of VEGFR2 antagonism and fibrosis/EndoMT inhibition was more effective than either individual treatment in reducing CNV. Our data indicate that fibrosis and EndoMT are involved in the progression of CNV, are exacerbated by VEGFR2 inhibition, and could provide an explanation for the reduced efficacy of anti-VEGFA treatment over time.
Collapse
|
26
|
Jeske R, Albo J, Marzano M, Bejoy J, Li Y. Engineering Brain-Specific Pericytes from Human Pluripotent Stem Cells. TISSUE ENGINEERING. PART B, REVIEWS 2020; 26:367-382. [PMID: 32571167 PMCID: PMC7462039 DOI: 10.1089/ten.teb.2020.0091] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 06/19/2020] [Indexed: 02/06/2023]
Abstract
Pericytes (PCs) are a type of perivascular cells that surround endothelial cells of small blood vessels. In the brain, PCs show heterogeneity depending on their position within the vasculature. As a result, PC interactions with surrounding endothelial cells, astrocytes, and neuron cells play a key role in a wide array of neurovascular functions such as regulating blood-brain barrier (BBB) permeability, cerebral blood flow, and helping to facilitate the clearance of toxic cellular molecules. Therefore, a reliable method of engineering brain-specific PCs from human induced pluripotent stem cells (hiPSCs) is critical in neurodegenerative disease modeling. This review summarizes brain-specific PC differentiation of hiPSCs through mesoderm and neural crest induction. Key signaling pathways (platelet-derived growth factor-B [PDGF-B], transforming growth factor [TGF]-β, and Notch signaling) regulating PC function, PC interactions with adjacent cells, and PC differentiation from hiPSCs are also discussed. Specifically, PDGF-BB-platelet-derived growth factor receptor β signaling promotes PC cell survival, TGF-β signal transduction facilitates PC attachment to endothelial cells, and Notch signaling is critical in vascular development and arterial-venous specification. Furthermore, current challenges facing the use of hiPSC-derived PCs are discussed, and their ongoing uses in neurodegenerative disease modeling are identified. Further investigations into PCs and surrounding cell interactions are needed to characterize the roles of brain PCs in various neurodegenerative disorders. Impact statement This article summarizes the work related to brain-specific pericytes (PCs) derived from human pluripotent stem cells (hPSCs). In particular, key signaling pathways regulating PC function, PC interactions with adjacent cells, and PC differentiation from hPSCs were discussed. Furthermore, current challenges facing the use of hPSC-derived PCs were identified, and their ongoing uses in neurodegenerative disease modeling were discussed. The review highlights the important role of cell-cell interactions in blood-brain barrier (BBB) models and neurodegeneration. The summarized findings are significant for establishing pluripotent stem cell-based BBB models toward the applications in drug screening and disease modeling.
Collapse
Affiliation(s)
- Richard Jeske
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, USA
| | - Jonathan Albo
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, USA
| | - Mark Marzano
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, USA
| | - Julie Bejoy
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, USA
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, USA
| |
Collapse
|
27
|
Zheng Z, Chopp M, Chen J. Multifaceted roles of pericytes in central nervous system homeostasis and disease. J Cereb Blood Flow Metab 2020; 40:1381-1401. [PMID: 32208803 PMCID: PMC7308511 DOI: 10.1177/0271678x20911331] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Pericytes, the mural cells surrounding microcirculation, are gaining increasing attention for their roles in health and disease of the central nervous system (CNS). As an essential part of the neurovascular unit (NVU), pericytes are actively engaged in interactions with neighboring cells and work in synergy with them to maintain homeostasis of the CNS, such as maintaining the blood-brain barrier (BBB), regulating cerebral blood flow (CBF) and the glymphatic system as well as mediating immune responses. However, the dysfunction of pericytes may contribute to the progression of various pathologies. In this review, we discuss: (1) origin of pericytes and different pericyte markers; (2) interactions of pericytes with endothelial cells (ECs), astrocytes, microglia, oligodendrocytes, and neurons; (3) physiological roles of pericytes in the CNS; (4) effects of pericytes in different CNS diseases; (5) relationship of pericytes with extracellular vesicles (EVs) and microRNAs (miRs); (6) recent advances in pericytes studies and future perspective.
Collapse
Affiliation(s)
- Zhitong Zheng
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA.,Department of Physics, Oakland University, Rochester, MI, USA
| | - Jieli Chen
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| |
Collapse
|
28
|
Rao M, Oh K, Moffitt R, Thompson P, Li J, Liu J, Sasson A, Georgakis G, Kim J, Choi M, Powers S. Comparative single-cell RNA sequencing (scRNA-seq) reveals liver metastasis-specific targets in a patient with small intestinal neuroendocrine cancer. Cold Spring Harb Mol Case Stud 2020; 6:mcs.a004978. [PMID: 32054662 PMCID: PMC7133744 DOI: 10.1101/mcs.a004978] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 02/03/2020] [Indexed: 01/08/2023] Open
Abstract
Genomic analysis of a patient's tumor is the cornerstone of precision oncology, but it does not address whether metastases should be treated differently. Here we tested whether comparative single-cell RNA sequencing (scRNA-seq) of a primary small intestinal neuroendocrine tumor to a matched liver metastasis could guide the treatment of a patient's metastatic disease. Following surgery, the patient was put on maintenance treatment with a somatostatin analog. However, the scRNA-seq analysis revealed that the neuroendocrine epithelial cells in the liver metastasis were less differentiated and expressed relatively little SSTR2, the predominant somatostatin receptor. There were also differences in the tumor microenvironments. RNA expression of vascular endothelial growth factors was higher in the primary tumor cells, reflected by an increased number of endothelial cells. Interestingly, vascular expression of the major VEGF receptors was considerably higher in the liver metastasis, indicating that the metastatic vasculature may be primed for expansion and susceptible to treatment with angiogenesis inhibitors. The patient eventually progressed on Sandostatin, and although consideration was given to adding an angiogenesis inhibitor to her regimen, her disease progression involved non-liver metastases that had not been characterized. Although in this specific case comparative scRNA-seq did not alter treatment, its potential to help guide therapy of metastatic disease was clearly demonstrated.
Collapse
Affiliation(s)
- Manisha Rao
- Department of Pathology, Stony Brook University, Stony Brook, New York 11794, USA
| | - Ki Oh
- Department of Medicine, Stony Brook University, Stony Brook, New York 11794, USA
| | - Richard Moffitt
- Department of Pathology, Stony Brook University, Stony Brook, New York 11794, USA.,Department of Biomedical Informatics, Stony Brook University, Stony Brook, New York 11794, USA
| | - Patricia Thompson
- Department of Pathology, Stony Brook University, Stony Brook, New York 11794, USA
| | - Jinyu Li
- Department of Pathology, Stony Brook University, Stony Brook, New York 11794, USA
| | - Jingxuan Liu
- Department of Pathology, Stony Brook University, Stony Brook, New York 11794, USA
| | - Aaron Sasson
- Department of Medicine, Stony Brook University, Stony Brook, New York 11794, USA
| | - George Georgakis
- Department of Medicine, Stony Brook University, Stony Brook, New York 11794, USA
| | - Joseph Kim
- Department of Medicine, Stony Brook University, Stony Brook, New York 11794, USA
| | - Minsig Choi
- Department of Medicine, Stony Brook University, Stony Brook, New York 11794, USA
| | - Scott Powers
- Department of Pathology, Stony Brook University, Stony Brook, New York 11794, USA
| |
Collapse
|
29
|
Yu F, Kumar NDS, Foo LC, Ng SH, Hunziker W, Choudhury D. A pump-free tricellular blood-brain barrier on-a-chip model to understand barrier property and evaluate drug response. Biotechnol Bioeng 2020; 117:1127-1136. [PMID: 31885078 DOI: 10.1002/bit.27260] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 12/06/2019] [Accepted: 12/22/2019] [Indexed: 12/18/2022]
Abstract
Disruption of the blood-brain barrier (BBB) leads to various neurovascular diseases. Development of therapeutics required to cross the BBB is difficult due to a lack of relevant in vitro models. We have developed a three-dimensional (3D) microfluidic BBB chip (BBBC) to study cell interactions in the brain microvasculature and to test drug candidates of neurovascular diseases. We isolated primary brain microvascular endothelial cells (ECs), pericytes, and astrocytes from neonatal rats and cocultured them in the BBBC. To mimic the 3D in vivo BBB structure, we used type I collagen hydrogel to pattern the microchannel via viscous finger patterning technique to create a matrix. ECs, astrocytes, and pericytes were cocultured in the collagen matrix. The fluid flow in the BBBC was controlled by a pump-free strategy utilizing gravity as driving force and resistance in a paper-based flow resistor. The primary cells cultured in the BBBC expressed high levels of junction proteins and formed a tight endothelial barrier layer. Addition of tumor necrosis factor alpha to recapitulate neuroinflammatory conditions compromised the BBB functionality. To mitigate the neuroinflammatory stimulus, we treated the BBB model with the glucocorticoid drug dexamethasone, and observed protection of the BBB. This BBBC represents a new simple, cost-effective, and scalable in vitro platform for validating therapeutic drugs targeting neuroinflammatory conditions.
Collapse
Affiliation(s)
- Fang Yu
- Bio-Manufacturing Group, Singapore Institute of Manufacturing Technology (SIMTech), A*STAR, Singapore, Singapore
| | - Nivasini D/O Selva Kumar
- Epithelial Cell Biology Laboratory, Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore
| | - Lynette C Foo
- Epithelial Cell Biology Laboratory, Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore
| | - Sum Huan Ng
- Bio-Manufacturing Group, Singapore Institute of Manufacturing Technology (SIMTech), A*STAR, Singapore, Singapore
| | - Walter Hunziker
- Epithelial Cell Biology Laboratory, Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Deepak Choudhury
- Bio-Manufacturing Group, Singapore Institute of Manufacturing Technology (SIMTech), A*STAR, Singapore, Singapore
| |
Collapse
|
30
|
Nakagomi T, Takagi T, Beppu M, Yoshimura S, Matsuyama T. Neural regeneration by regionally induced stem cells within post-stroke brains: Novel therapy perspectives for stroke patients. World J Stem Cells 2019; 11:452-463. [PMID: 31523366 PMCID: PMC6716084 DOI: 10.4252/wjsc.v11.i8.452] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 07/04/2019] [Accepted: 07/16/2019] [Indexed: 02/06/2023] Open
Abstract
Ischemic stroke is a critical disease which causes serious neurological functional loss such as paresis. Hope for novel therapies is based on the increasing evidence of the presence of stem cell populations in the central nervous system (CNS) and the development of stem-cell-based therapies for stroke patients. Although mesenchymal stem cells (MSCs) represented initially a promising cell source, only a few transplanted MSCs were present near the injured areas of the CNS. Thus, regional stem cells that are present and/or induced in the CNS may be ideal when considering a treatment following ischemic stroke. In this context, we have recently showed that injury/ischemia-induced neural stem/progenitor cells (iNSPCs) and injury/ischemia-induced multipotent stem cells (iSCs) are present within post-stroke human brains and post-stroke mouse brains. This indicates that iNSPCs/iSCs could be developed for clinical applications treating patients with stroke. The present study introduces the traits of mouse and human iNSPCs, with a focus on the future perspective for CNS regenerative therapies using novel iNSPCs/iSCs.
Collapse
Affiliation(s)
- Takayuki Nakagomi
- Institute for Advanced Medical Sciences, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
- Department of Therapeutic Progress in Brain Diseases, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Toshinori Takagi
- Department of Neurosurgery, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Mikiya Beppu
- Department of Neurosurgery, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Shinichi Yoshimura
- Department of Neurosurgery, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Tomohiro Matsuyama
- Department of Therapeutic Progress in Brain Diseases, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| |
Collapse
|
31
|
Bertrand L, Cho HJ, Toborek M. Blood-brain barrier pericytes as a target for HIV-1 infection. Brain 2019; 142:502-511. [PMID: 30668645 PMCID: PMC6391611 DOI: 10.1093/brain/awy339] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 10/19/2018] [Accepted: 11/06/2018] [Indexed: 12/11/2022] Open
Abstract
Pericytes are multifunctional cells wrapped around endothelial cells via cytoplasmic processes that extend along the abluminal surface of the endothelium. The interactions between endothelial cells and pericytes of the blood-brain barrier are necessary for proper formation, development, stabilization, and maintenance of the blood-brain barrier. Blood-brain barrier pericytes regulate paracellular flow between cells, transendothelial fluid transport, maintain optimal chemical composition of the surrounding microenvironment, and protect endothelial cells from potential harmful substances. Thus, dysfunction or loss of blood-brain barrier pericytes is an important factor in the pathogenesis of several diseases that are associated with microvascular instability. Importantly, recent research indicates that blood-brain barrier pericytes can be a target of HIV-1 infection able to support productive HIV-1 replication. In addition, blood-brain barrier pericytes are prone to establish a latent infection, which can be reactivated by a mixture of histone deacetylase inhibitors in combination with TNF. HIV-1 infection of blood-brain barrier pericytes has been confirmed in a mouse model of HIV-1 infection and in human post-mortem samples of HIV-1-infected brains. Overall, recent evidence indicates that blood-brain barrier pericytes can be a previously unrecognized HIV-1 target and reservoir in the brain.
Collapse
Affiliation(s)
- Luc Bertrand
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Hyung Joon Cho
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA,Correspondence to: Michal Toborek Department of Biochemistry and Molecular Biology University of Miami School of Medicine Gautier Bldg., Room 528 1011 NW 15th Street Miami, FL 33136, USA E-mail:
| |
Collapse
|
32
|
Bennett C, Mohammed F, Álvarez-Ciara A, Nguyen MA, Dietrich WD, Rajguru SM, Streit WJ, Prasad A. Neuroinflammation, oxidative stress, and blood-brain barrier (BBB) disruption in acute Utah electrode array implants and the effect of deferoxamine as an iron chelator on acute foreign body response. Biomaterials 2019; 188:144-159. [PMID: 30343257 PMCID: PMC6300159 DOI: 10.1016/j.biomaterials.2018.09.040] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 09/20/2018] [Accepted: 09/24/2018] [Indexed: 02/06/2023]
Abstract
The use of intracortical microelectrode arrays has gained significant attention in being able to help restore function in paralysis patients and study the brain in various neurological disorders. Electrode implantation in the cortex causes vasculature or blood-brain barrier (BBB) disruption and thus elicits a foreign body response (FBR) that results in chronic inflammation and may lead to poor electrode performance. In this study, a comprehensive insight into the acute molecular mechanisms occurring at the Utah electrode array-tissue interface is provided to understand the oxidative stress, neuroinflammation, and neurovascular unit (astrocytes, pericytes, and endothelial cells) disruption that occurs following microelectrode implantation. Quantitative real time polymerase chain reaction (qRT-PCR) was used to quantify the gene expression at acute time-points of 48-hr, 72-hr, and 7-days for factors mediating oxidative stress, inflammation, and BBB disruption in rats implanted with a non-functional 4 × 4 Utah array in the somatosensory cortex. During vascular disruption, free iron released into the brain parenchyma can exacerbate the FBR, leading to oxidative stress and thus further contributing to BBB degradation. To reduce the free iron released into the brain tissue, the effects of an iron chelator, deferoxamine mesylate (DFX), was also evaluated.
Collapse
Affiliation(s)
- Cassie Bennett
- Department of Biomedical Engineering, University of Miami, FL, USA
| | - Farrah Mohammed
- Department of Biomedical Engineering, University of Miami, FL, USA
| | | | | | | | - Suhrud M Rajguru
- Department of Biomedical Engineering, University of Miami, FL, USA
| | | | - Abhishek Prasad
- Department of Biomedical Engineering, University of Miami, FL, USA.
| |
Collapse
|
33
|
Lee LL, Chintalgattu V. Pericytes in the Heart. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1122:187-210. [PMID: 30937870 DOI: 10.1007/978-3-030-11093-2_11] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Mural cells known as pericytes envelop the endothelial layer of microvessels throughout the body and have been described to have tissue-specific functions. Cardiac pericytes are abundantly found in the heart, but they are relatively understudied. Currently, their importance is emerging in cardiovascular homeostasis and dysfunction due to their pleiotropism. They are known to play key roles in vascular tone and vascular integrity as well as angiogenesis. However, their dysfunctional presence and/or absence is critical in the mechanisms that lead to cardiac pathologies such as myocardial infarction, fibrosis, and thrombosis. Moreover, they are targeted as a therapeutic potential due to their mesenchymal properties that could allow them to repair and regenerate a damaged heart. They are also sought after as a cell-based therapy based on their healing potential in preclinical studies of animal models of myocardial infarction. Therefore, recognizing the importance of cardiac pericytes and understanding their biology will lead to new therapeutic concepts.
Collapse
Affiliation(s)
- Linda L Lee
- Department of CardioMetabolic Disorders, Amgen Research and Discovery, Amgen Inc., South San Francisco, CA, USA
| | - Vishnu Chintalgattu
- Department of CardioMetabolic Disorders, Amgen Research and Discovery, Amgen Inc., South San Francisco, CA, USA.
| |
Collapse
|
34
|
Schwartz SM, Virmani R, Majesky MW. An update on clonality: what smooth muscle cell type makes up the atherosclerotic plaque? F1000Res 2018; 7:F1000 Faculty Rev-1969. [PMID: 30613386 PMCID: PMC6305222 DOI: 10.12688/f1000research.15994.1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/06/2018] [Indexed: 12/13/2022] Open
Abstract
Almost 50 years ago, Earl Benditt and his son John described the clonality of the atherosclerotic plaque. This led Benditt to propose that the atherosclerotic lesion was a smooth muscle neoplasm, similar to the leiomyomata seen in the uterus of most women. Although the observation of clonality has been confirmed many times, interest in the idea that atherosclerosis might be a form of neoplasia waned because of the clinical success of treatments for hyperlipemia and because animal models have made great progress in understanding how lipid accumulates in the plaque and may lead to plaque rupture. Four advances have made it important to reconsider Benditt's observations. First, we now know that clonality is a property of normal tissue development. Second, this is even true in the vessel wall, where we now know that formation of clonal patches in that wall is part of the development of smooth muscle cells that make up the tunica media of arteries. Third, we know that the intima, the "soil" for development of the human atherosclerotic lesion, develops before the fatty lesions appear. Fourth, while the cells comprising this intima have been called "smooth muscle cells", we do not have a clear definition of cell type nor do we know if the initial accumulation is clonal. As a result, Benditt's hypothesis needs to be revisited in terms of changes in how we define smooth muscle cells and the quite distinct developmental origins of the cells that comprise the muscular coats of all arterial walls. Finally, since clonality of the lesions is real, the obvious questions are do these human tumors precede the development of atherosclerosis, how do the clones develop, what cell type gives rise to the clones, and in what ways do the clones provide the soil for development and natural history of atherosclerosis?
Collapse
Affiliation(s)
| | - Renu Virmani
- CV Path Institute, Gaithersberg, Maryland, 20878, USA
| | - Mark W. Majesky
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Hospital Research Institute, Seattle, WA, 98112, USA
| |
Collapse
|
35
|
Wang D, Xu Y, Feng L, Yin P, Song SS, Wu F, Yan P, Liang Z. RGS5 decreases the proliferation of human ovarian carcinoma‑derived primary endothelial cells through the MAPK/ERK signaling pathway in hypoxia. Oncol Rep 2018; 41:165-177. [PMID: 30365142 PMCID: PMC6278583 DOI: 10.3892/or.2018.6811] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 09/17/2018] [Indexed: 12/20/2022] Open
Abstract
Regulator of G-protein signaling 5 (RGS5), a tissue-specific signal-regulating molecule, plays a key role in the development of the vasculature. It was recently found that RGS5 is abundantly expressed in epithelial ovarian cancer (EOC) compared with the normal ovaries. However, the distribution of RGS5 in EOC and its significance require further investigation. The aim of the present study was to investigate the expression of RGS5 in EOC, as well as its association with cancer differentiation, metastasis and clinicopathological parameters. Immunohistochemistry (IHC), western blotting, RT-PCR, wound-healing, cell proliferation and flow cytometric assays were the methods used in the present study. RGS5 was highly expressed in the cytoplasm of ovarian carcinoma cells and in microvascular structures. The expression of RGS5 in EOC was negatively associated with peritoneal metastasis (P=0.004), but it was not found to be associated with age, tumor size, clinical stage or lymph node metastasis (P>0.05). EOC patients with high RGS5 expression had a prolonged progression-free survival (72.34±8.41 vs. 43.56±5.41 months, P<0.001). High expression of RGS5 was correlated with significantly lower microvascular density (MVD) as indicated by the expression of CD34, whereas the opposite was observed in tissues with low RGS5 expression (P<0.05). Hypoxia increased RGS5 expression in ovarian carcinoma-derived endothelial cells (ODMECs), whereas the proliferative capacity of ODMECs exhibited a significant increase following RNAi-mediated reduction of RGS5 expression. These data indicated that RGS5 plays a key role in angiogenesis in ovarian carcinoma. In addition, RGS5 downregulated the expression of the downstream proteins CDC25A, CDK2 and cyclin E, which are mediated by the mitogen-activated protein kinase/extracellular signal-regulated kinase pathway, causing ODMEC arrest in the G1 phase of the cell cycle under hypoxic conditions. Collectively, our data indicated that RGS5 is crucial for the occurrence and development of ovarian cancer, and that RGS5 and its signaling pathway may serve as anti-angiogenesis targets for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Dan Wang
- Department of Obstetrics and Gynecology, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Yan Xu
- 77103rd troops, PLA, Chongqing 400038, P.R. China
| | - Lu Feng
- Department of Obstetrics and Gynecology, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Pin Yin
- Department of Obstetrics and Gynecology, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Shuang Shuang Song
- Department of Geriatrics, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Feng Wu
- Department of Pathology, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Ping Yan
- Department of Obstetrics and Gynecology, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Zhiqing Liang
- Department of Obstetrics and Gynecology, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| |
Collapse
|
36
|
Kolinko Y, Kralickova M, Tonar Z. The impact of pericytes on the brain and approaches for their morphological analysis. J Chem Neuroanat 2018; 91:35-45. [DOI: 10.1016/j.jchemneu.2018.04.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 04/10/2018] [Accepted: 04/15/2018] [Indexed: 12/15/2022]
|
37
|
Squires KE, Montañez-Miranda C, Pandya RR, Torres MP, Hepler JR. Genetic Analysis of Rare Human Variants of Regulators of G Protein Signaling Proteins and Their Role in Human Physiology and Disease. Pharmacol Rev 2018; 70:446-474. [PMID: 29871944 PMCID: PMC5989036 DOI: 10.1124/pr.117.015354] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Regulators of G protein signaling (RGS) proteins modulate the physiologic actions of many neurotransmitters, hormones, and other signaling molecules. Human RGS proteins comprise a family of 20 canonical proteins that bind directly to G protein-coupled receptors/G protein complexes to limit the lifetime of their signaling events, which regulate all aspects of cell and organ physiology. Genetic variations account for diverse human traits and individual predispositions to disease. RGS proteins contribute to many complex polygenic human traits and pathologies such as hypertension, atherosclerosis, schizophrenia, depression, addiction, cancers, and many others. Recent analysis indicates that most human diseases are due to extremely rare genetic variants. In this study, we summarize physiologic roles for RGS proteins and links to human diseases/traits and report rare variants found within each human RGS protein exome sequence derived from global population studies. Each RGS sequence is analyzed using recently described bioinformatics and proteomic tools for measures of missense tolerance ratio paired with combined annotation-dependent depletion scores, and protein post-translational modification (PTM) alignment cluster analysis. We highlight selected variants within the well-studied RGS domain that likely disrupt RGS protein functions and provide comprehensive variant and PTM data for each RGS protein for future study. We propose that rare variants in functionally sensitive regions of RGS proteins confer profound change-of-function phenotypes that may contribute, in newly appreciated ways, to complex human diseases and/or traits. This information provides investigators with a valuable database to explore variation in RGS protein function, and for targeting RGS proteins as future therapeutic targets.
Collapse
Affiliation(s)
- Katherine E Squires
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.)
| | - Carolina Montañez-Miranda
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.)
| | - Rushika R Pandya
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.)
| | - Matthew P Torres
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.)
| | - John R Hepler
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.)
| |
Collapse
|
38
|
Yamazaki T, Mukouyama YS. Tissue Specific Origin, Development, and Pathological Perspectives of Pericytes. Front Cardiovasc Med 2018; 5:78. [PMID: 29998128 PMCID: PMC6030356 DOI: 10.3389/fcvm.2018.00078] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Accepted: 06/07/2018] [Indexed: 01/09/2023] Open
Abstract
Pericytes are mural cells surrounding blood vessels, adjacent to endothelial cells. Pericytes play critical roles in maturation and maintenance of vascular branching morphogenesis. In the central nervous system (CNS), pericytes are necessary for the formation and regulation of the blood-brain barrier (BBB) and pericyte deficiency accompanies CNS diseases including multiple sclerosis, diabetic retinopathy, neonatal intraventricular hemorrhage, and neurodegenerative disorders. Despite the importance of pericytes, their developmental origins and phenotypic diversity remain incompletely understood. Pericytes express multiple markers and the origin of pericytes differs by tissue, which may cause difficulty for the identification and understanding of the ontogeny of pericytes. Also, pericytes have the potential to give rise to different tissues in vitro but this is not clear in vivo. These studies indicate that pericytes are heterogeneous in a tissue- and context- dependent manner. This short review focuses on recent studies about identification of pericytes, heterogeneous origin of pericytes during development and in adults, and the differentiation capacity of pericytes, and pericytes in pathological settings.
Collapse
Affiliation(s)
- Tomoko Yamazaki
- Laboratory of Stem Cell and Neuro-Vascular Biology, Genetics and Developmental Biology Center, National Heart, Lung, and Blood Institute, Bethesda, MD, United States.,Robert W. Franz Cancer Center, Providence Portland Medical Center, Earle A. Chiles Research Institute, Portland, OR, United States
| | - Yoh-Suke Mukouyama
- Laboratory of Stem Cell and Neuro-Vascular Biology, Genetics and Developmental Biology Center, National Heart, Lung, and Blood Institute, Bethesda, MD, United States
| |
Collapse
|
39
|
Padel T, Roth M, Gaceb A, Li JY, Björkqvist M, Paul G. Brain pericyte activation occurs early in Huntington's disease. Exp Neurol 2018; 305:139-150. [PMID: 29630897 DOI: 10.1016/j.expneurol.2018.03.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/03/2018] [Accepted: 03/29/2018] [Indexed: 02/03/2023]
Abstract
Microvascular changes have recently been described for several neurodegenerative disorders, including Huntington's disease (HD). HD is characterized by a progressive neuronal cell loss due to a mutation in the Huntingtin gene. However, the temporal and spatial microvascular alterations in HD remain unclear. Also, knowledge on the implication of pericytes in HD pathology is still sparse and existing findings are contradictory. Here we examine alterations in brain pericytes in the R6/2 mouse model of HD and in human post mortem HD brain sections. To specifically track activated pericytes, we crossbred R6/2 mice with transgenic mice expressing the Green fluorescent protein gene under the Regulator of G-protein signaling 5 (Rgs5) promoter. We demonstrate an increase in activated pericytes in the R6/2 brain and in post mortem HD brain tissue. Importantly, pericyte changes are already detected before striatal neuronal cell loss, weight loss or behavioural deficits occur in R6/2 mice. This is associated with vascular alterations, whereby striatal changes precede cortical changes. Our findings suggest that pericyte activation may be one of the initial steps contributing to the observed vascular modifications in HD. Thus, pericytes may constitute an important target to address early microvascular changes contributing to disease progression in HD.
Collapse
Affiliation(s)
- Thomas Padel
- Translational Neurology group, Department of Clinical Science, Wallenberg Neuroscience Center, Wallenberg Centre for Molecular Medicine at Lund University, Lund University, 22184 Lund, Sweden
| | - Michaela Roth
- Translational Neurology group, Department of Clinical Science, Wallenberg Neuroscience Center, Wallenberg Centre for Molecular Medicine at Lund University, Lund University, 22184 Lund, Sweden
| | - Abderahim Gaceb
- Translational Neurology group, Department of Clinical Science, Wallenberg Neuroscience Center, Wallenberg Centre for Molecular Medicine at Lund University, Lund University, 22184 Lund, Sweden
| | - Jia-Yi Li
- Neural Plasticity and Repair Unit, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, 22184 Lund, Sweden
| | - Maria Björkqvist
- Biomarkers in Brain Disease, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, 22184 Lund, Sweden
| | - Gesine Paul
- Translational Neurology group, Department of Clinical Science, Wallenberg Neuroscience Center, Wallenberg Centre for Molecular Medicine at Lund University, Lund University, 22184 Lund, Sweden; Department of Neurology, Scania University Hospital, 22185 Lund, Sweden.
| |
Collapse
|
40
|
Tsukada T, Yoshida S, Kito K, Fujiwara K, Yako H, Horiguchi K, Isowa Y, Yashiro T, Kato T, Kato Y. TGFβ signaling reinforces pericyte properties of the non-endocrine mouse pituitary cell line TtT/GF. Cell Tissue Res 2017; 371:339-350. [DOI: 10.1007/s00441-017-2758-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 11/19/2017] [Indexed: 01/11/2023]
|
41
|
Bartoszewski R, Serocki M, Janaszak-Jasiecka A, Bartoszewska S, Kochan-Jamrozy K, Piotrowski A, Króliczewski J, Collawn JF. miR-200b downregulates Kruppel Like Factor 2 (KLF2) during acute hypoxia in human endothelial cells. Eur J Cell Biol 2017; 96:758-766. [PMID: 29042072 PMCID: PMC5677561 DOI: 10.1016/j.ejcb.2017.10.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 10/01/2017] [Accepted: 10/11/2017] [Indexed: 01/03/2023] Open
Abstract
The role of microRNAs in controlling angiogenesis is recognized as a promising therapeutic target in both cancer and cardiovascular disorders. However, understanding a miRNA's pleiotropic effects on angiogenesis is a limiting factor for these types of therapeutic approaches. Using genome-wide next-generation sequencing, we examined the role of an antiangiogenic miRNA, miR-200b, in primary human endothelial cells. The results indicate that miR-200b has complex effects on hypoxia-induced angiogenesis in human endothelia and importantly, that many of the reported miR-200b effects using miRNA overexpression may not be representative of the physiological role of this miRNA. We also identified the antiangiogenic KLF2 gene as a novel target of miR-200b. Our studies indicate that the physiological changes in miR-200b levels during acute hypoxia may actually have a proangiogenic effect through Klf2 downregulation and subsequent stabilization of HIF-1 signaling. Moreover, we provide a viable approach for differentiating direct from indirect miRNA effects in order to untangle the complexity of individual miRNA networks.
Collapse
Affiliation(s)
- Rafal Bartoszewski
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland.
| | - Marcin Serocki
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| | - Anna Janaszak-Jasiecka
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| | - Sylwia Bartoszewska
- Department of Inorganic Chemistry, Medical University of Gdansk, Gdansk, Poland
| | - Kinga Kochan-Jamrozy
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| | - Arkadiusz Piotrowski
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| | - Jarosław Króliczewski
- Laboratory of Chemical Biology, Faculty of Biotechnology, University of Wroclaw, Poland
| | - James F Collawn
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, USA
| |
Collapse
|
42
|
Gaskill CF, Carrier EJ, Kropski JA, Bloodworth NC, Menon S, Foronjy RF, Taketo MM, Hong CC, Austin ED, West JD, Means AL, Loyd JE, Merryman WD, Hemnes AR, De Langhe S, Blackwell TS, Klemm DJ, Majka SM. Disruption of lineage specification in adult pulmonary mesenchymal progenitor cells promotes microvascular dysfunction. J Clin Invest 2017; 127:2262-2276. [PMID: 28463231 DOI: 10.1172/jci88629] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 03/02/2017] [Indexed: 01/04/2023] Open
Abstract
Pulmonary vascular disease is characterized by remodeling and loss of microvessels and is typically attributed to pathological responses in vascular endothelium or abnormal smooth muscle cell phenotypes. We have challenged this understanding by defining an adult pulmonary mesenchymal progenitor cell (MPC) that regulates both microvascular function and angiogenesis. The current understanding of adult MPCs and their roles in homeostasis versus disease has been limited by a lack of genetic markers with which to lineage label multipotent mesenchyme and trace the differentiation of these MPCs into vascular lineages. Here, we have shown that lineage-labeled lung MPCs expressing the ATP-binding cassette protein ABCG2 (ABCG2+) are pericyte progenitors that participate in microvascular homeostasis as well as adaptive angiogenesis. Activation of Wnt/β-catenin signaling, either autonomously or downstream of decreased BMP receptor signaling, enhanced ABCG2+ MPC proliferation but suppressed MPC differentiation into a functional pericyte lineage. Thus, enhanced Wnt/β-catenin signaling in ABCG2+ MPCs drives a phenotype of persistent microvascular dysfunction, abnormal angiogenesis, and subsequent exacerbation of bleomycin-induced fibrosis. ABCG2+ MPCs may, therefore, account in part for the aberrant microvessel function and remodeling that are associated with chronic lung diseases.
Collapse
Affiliation(s)
- Christa F Gaskill
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine or Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee USA
| | - Erica J Carrier
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine or Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee USA
| | - Jonathan A Kropski
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine or Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee USA
| | | | - Swapna Menon
- Pulmonary Vascular Research Institute, Kochi, and AnalyzeDat Consulting Services, Kerala, India
| | - Robert F Foronjy
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, SUNY Downstate Medical Center, Brooklyn, New York, USA
| | | | - Charles C Hong
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine or Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee USA.,Department of Pathology and Laboratory Medicine or Department of Medicine, Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | | | - James D West
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine or Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee USA
| | - Anna L Means
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - James E Loyd
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine or Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee USA
| | - W David Merryman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee USA
| | - Anna R Hemnes
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine or Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee USA
| | | | - Timothy S Blackwell
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine or Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee USA
| | - Dwight J Klemm
- Department of Medicine, Pulmonary and Critical Care Medicine, Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado, Aurora, Colorado, USA.,Geriatric Research Education and Clinical Center, Eastern Colorado Health Care System, Denver, Colorado, USA
| | - Susan M Majka
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine or Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee USA.,Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
43
|
Jaffe GJ, Ciulla TA, Ciardella AP, Devin F, Dugel PU, Eandi CM, Masonson H, Monés J, Pearlman JA, Quaranta-El Maftouhi M, Ricci F, Westby K, Patel SC. Dual Antagonism of PDGF and VEGF in Neovascular Age-Related Macular Degeneration. Ophthalmology 2017; 124:224-234. [DOI: 10.1016/j.ophtha.2016.10.010] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 10/06/2016] [Accepted: 10/08/2016] [Indexed: 01/20/2023] Open
|
44
|
Abdeen AA, Lee J, Li Y, Kilian KA. Cytoskeletal Priming of Mesenchymal Stem Cells to a Medicinal Phenotype. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2017. [DOI: 10.1007/s40883-016-0021-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
45
|
Yuan K, Shao NY, Hennigs JK, Discipulo M, Orcholski ME, Shamskhou E, Richter A, Hu X, Wu JC, de Jesus Perez VA. Increased Pyruvate Dehydrogenase Kinase 4 Expression in Lung Pericytes Is Associated with Reduced Endothelial-Pericyte Interactions and Small Vessel Loss in Pulmonary Arterial Hypertension. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2500-14. [PMID: 27456128 DOI: 10.1016/j.ajpath.2016.05.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 04/26/2016] [Accepted: 05/25/2016] [Indexed: 12/23/2022]
Abstract
Reduced endothelial-pericyte interactions are linked to progressive small vessel loss in pulmonary arterial hypertension (PAH), but the molecular mechanisms underlying this disease remain poorly understood. To identify relevant gene candidates associated with aberrant pericyte behavior, we performed a transcriptome analysis of patient-derived donor control and PAH lung pericytes followed by functional genomics analysis. Compared with donor control cells, PAH pericytes had significant enrichment of genes involved in various metabolic processes, the top hit being PDK4, a gene coding for an enzyme that suppresses mitochondrial activity in favor of glycolysis. Given reports that link reduced mitochondrial activity with increased PAH cell proliferation, we hypothesized that increased PDK4 is associated with PAH pericyte hyperproliferation and reduced endothelial-pericyte interactions. We found that PDK4 gene and protein expression was significantly elevated in PAH pericytes and correlated with reduced mitochondrial metabolism, higher rates of glycolysis, and hyperproliferation. Importantly, reducing PDK4 levels restored mitochondrial metabolism, reduced cell proliferation, and improved endothelial-pericyte interactions. To our knowledge, this is the first study that documents significant differences in gene expression between human donor control and PAH lung pericytes and the link between mitochondrial dysfunction and aberrant endothelial-pericyte interactions in PAH. Comprehensive characterization of these candidate genes could provide novel therapeutic targets to improve endothelial-pericyte interactions and prevent small vessel loss in PAH.
Collapse
Affiliation(s)
- Ke Yuan
- Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, California; Stanford Cardiovascular Institute, Stanford University, Stanford, California
| | - Ning-Yi Shao
- Stanford Cardiovascular Institute, Stanford University, Stanford, California; Division of Cardiology, Stanford University, Stanford, California
| | - Jan K Hennigs
- Stanford Cardiovascular Institute, Stanford University, Stanford, California; Department of Pediatrics, Stanford University, Stanford, California
| | - Marielle Discipulo
- Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, California
| | - Mark E Orcholski
- Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, California; Stanford Cardiovascular Institute, Stanford University, Stanford, California
| | - Elya Shamskhou
- Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, California; Stanford Cardiovascular Institute, Stanford University, Stanford, California
| | - Alice Richter
- Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, California
| | - Xinqian Hu
- Department of Genetics, Stanford University, Stanford, California
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University, Stanford, California; Division of Cardiology, Stanford University, Stanford, California
| | - Vinicio A de Jesus Perez
- Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, California; Stanford Cardiovascular Institute, Stanford University, Stanford, California.
| |
Collapse
|
46
|
Shen J, Shrestha S, Yen YH, Scott MA, Soo C, Ting K, Peault B, Dry SM, James AW. The pericyte antigen RGS5 in perivascular soft tissue tumors. Hum Pathol 2015; 47:121-31. [PMID: 26558691 DOI: 10.1016/j.humpath.2015.09.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 09/07/2015] [Accepted: 09/10/2015] [Indexed: 12/12/2022]
Abstract
Perivascular soft tissue tumors are relatively uncommon neoplasms of unclear lineage of differentiation, although most are presumed to originate from or differentiate to pericytes or a modified perivascular cell. Among these, glomus tumor, myopericytoma, and angioleiomyoma share a spectrum of histologic findings and a perivascular growth pattern. In contrast, solitary fibrous tumor was once hypothesized to have pericytic differentiation--although little bona fide evidence of pericytic differentiation exists. Likewise the perivascular epithelioid cell tumor (PEComa) family shares a perivascular growth pattern, but with distinctive dual myoid-melanocytic differentiation. RGS5, regulator of G-protein signaling 5, is a novel pericyte antigen with increasing use in animal models. Here, we describe the immunohistochemical expression patterns of RGS5 across perivascular soft tissue tumors, including glomus tumor (n = 6), malignant glomus tumor (n = 4), myopericytoma (n = 3), angioleiomyoma (n = 9), myofibroma (n = 4), solitary fibrous tumor (n = 10), and PEComa (n = 19). Immunohistochemical staining and semi-quantification was performed, and compared to αSMA (smooth muscle actin) expression. Results showed that glomus tumor (including malignant glomus tumor), myopericytoma, and angioleiomyoma shared a similar diffuse immunoreactivity for RGS5 and αSMA across all tumors examined. In contrast, myofibroma, solitary fibrous tumor and PEComa showed predominantly focal to absent RGS5 immunoreactivity. These findings further support a common pericytic lineage of differentiation in glomus tumors, myopericytoma and angioleiomyoma. The pericyte marker RGS5 may be of future clinical utility for the evaluation of pericytic differentiation in soft tissue tumors.
Collapse
Affiliation(s)
- Jia Shen
- School of Dentistry, University of California, Los Angeles, CA, 90095
| | - Swati Shrestha
- School of Dentistry, University of California, Los Angeles, CA, 90095; Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095
| | - Yu-Hsin Yen
- School of Dentistry, University of California, Los Angeles, CA, 90095; Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095
| | | | - Chia Soo
- Orthopedic Hospital Research Center, University of California, Los Angeles, CA, 90095; Department of Surgery, University of California, Los Angeles, Los Angeles, CA, 90095
| | - Kang Ting
- School of Dentistry, University of California, Los Angeles, CA, 90095
| | - Bruno Peault
- Orthopedic Hospital Research Center, University of California, Los Angeles, CA, 90095; Center for Regenerative Medicine, University of Edinburgh, Edinburgh, UK, EH16 4UU
| | - Sarah M Dry
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095
| | - Aaron W James
- School of Dentistry, University of California, Los Angeles, CA, 90095; Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095; Orthopedic Hospital Research Center, University of California, Los Angeles, CA, 90095.
| |
Collapse
|
47
|
Nakagomi T, Nakano-Doi A, Kawamura M, Matsuyama T. Do Vascular Pericytes Contribute to Neurovasculogenesis in the Central Nervous System as Multipotent Vascular Stem Cells? Stem Cells Dev 2015; 24:1730-9. [PMID: 25900222 DOI: 10.1089/scd.2015.0039] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Increasing evidence suggests that multipotent stem cells are harbored within a vascular niche inside various organs. Although a precise phenotype of resident vascular stem cells (VSCs) that can function as multipotent stem cells remains unclear, accumulating evidence shows that multipotent VSCs are likely vascular pericytes (PCs) that localize within blood vessels. These PCs are multipotent, possessing the ability to differentiate into various cell types, including vascular lineage cells. In addition, brain PCs are unique: They are derived from neural crest and can differentiate into neural lineage cells. Because PCs in the central nervous system (CNS) can contribute to both neurogenesis and vasculogenesis, they may mediate the reparative process of neurovascular units that are constructed by neural and vascular cells. Here, we describe the activity of PCs when viewed as multipotent VSCs, primarily regarding their neurogenic and vasculogenic potential in the CNS. We also discuss similarities between PCs and other candidates for multipotent VSCs, including perivascular mesenchymal stem cells, neural crest-derived stem cells, adventitial progenitor cells, and adipose-derived stem cells.
Collapse
Affiliation(s)
- Takayuki Nakagomi
- 1 Institute for Advanced Medical Sciences, Hyogo College of Medicine , Hyogo, Japan
| | - Akiko Nakano-Doi
- 1 Institute for Advanced Medical Sciences, Hyogo College of Medicine , Hyogo, Japan
| | - Miki Kawamura
- 1 Institute for Advanced Medical Sciences, Hyogo College of Medicine , Hyogo, Japan .,2 Department of Neurology, Osaka University Graduate School of Medicine , Osaka, Japan
| | - Tomohiro Matsuyama
- 1 Institute for Advanced Medical Sciences, Hyogo College of Medicine , Hyogo, Japan
| |
Collapse
|
48
|
Ganss R. Keeping the Balance Right. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 133:93-121. [DOI: 10.1016/bs.pmbts.2015.02.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
49
|
Marriott S, Baskir RS, Gaskill C, Menon S, Carrier EJ, Williams J, Talati M, Helm K, Alford CE, Kropski JA, Loyd J, Wheeler L, Johnson J, Austin E, Nozik-Grayck E, Meyrick B, West JD, Klemm DJ, Majka SM. ABCG2pos lung mesenchymal stem cells are a novel pericyte subpopulation that contributes to fibrotic remodeling. Am J Physiol Cell Physiol 2014; 307:C684-98. [PMID: 25122876 PMCID: PMC4200000 DOI: 10.1152/ajpcell.00114.2014] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 08/05/2014] [Indexed: 01/13/2023]
Abstract
Genesis of myofibroblasts is obligatory for the development of pathology in many adult lung diseases. Adult lung tissue contains a population of perivascular ABCG2(pos) mesenchymal stem cells (MSC) that are precursors of myofibroblasts and distinct from NG2 pericytes. We hypothesized that these MSC participate in deleterious remodeling associated with pulmonary fibrosis (PF) and associated hypertension (PH). To test this hypothesis, resident lung MSC were quantified in lung samples from control subjects and PF patients. ABCG2(pos) cell numbers were decreased in human PF and interstitial lung disease compared with control samples. Genetic labeling of lung MSC in mice enabled determination of terminal lineage and localization of ABCG2 cells following intratracheal administration of bleomycin to elicit fibrotic lung injury. Fourteen days following bleomycin injury enhanced green fluorescent protein (eGFP)-labeled lung MSC-derived cells were increased in number and localized to interstitial areas of fibrotic and microvessel remodeling. Finally, gene expression analysis was evaluated to define the response of MSC to bleomycin injury in vivo using ABCG2(pos) MSC isolated during the inflammatory phase postinjury and in vitro bleomycin or transforming growth factor-β1 (TGF-β1)-treated cells. MSC responded to bleomycin treatment in vivo with a profibrotic gene program that was not recapitulated in vitro with bleomycin treatment. However, TGF-β1 treatment induced the appearance of a profibrotic myofibroblast phenotype in vitro. Additionally, when exposed to the profibrotic stimulus, TGF-β1, ABCG2, and NG2 pericytes demonstrated distinct responses. Our data highlight ABCG2(pos) lung MSC as a novel cell population that contributes to detrimental myofibroblast-mediated remodeling during PF.
Collapse
Affiliation(s)
- Shennea Marriott
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University, Nashville, Tennesse
| | - Rubin S Baskir
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennesse
| | - Christa Gaskill
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University, Nashville, Tennesse
| | - Swapna Menon
- Pulmonary Vascular Research Institute Kochi and AnalyzeDat Consulting Services, Kerala, India
| | - Erica J Carrier
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University, Nashville, Tennesse
| | - Janice Williams
- Vanderbilt Ingram Cancer Center, Electron Microscopy-Cell Imaging Shared Resource, Vanderbilt University, Nashville, Tennessee
| | - Megha Talati
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University, Nashville, Tennesse
| | - Karen Helm
- Cancer Center Flow Cytometry Shared Resource, University of Colorado, Aurora, Colorado
| | - Catherine E Alford
- Department of Pathology and Laboratory Medicine, Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee
| | - Jonathan A Kropski
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University, Nashville, Tennesse
| | - James Loyd
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University, Nashville, Tennesse
| | - Lisa Wheeler
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University, Nashville, Tennesse
| | - Joyce Johnson
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee
| | - Eric Austin
- Department of Pediatrics, Vanderbilt University, Nashville, Tennessee
| | - Eva Nozik-Grayck
- Department of Pediatrics or Medicine, Pulmonary and Critical Care Medicine, Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado, Aurora, Colorado; and
| | - Barbara Meyrick
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University, Nashville, Tennesse
| | - James D West
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University, Nashville, Tennesse; Vanderbilt Pulmonary Circulation Center, Vanderbilt University, Nashville, Tennessee
| | - Dwight J Klemm
- Department of Pediatrics or Medicine, Pulmonary and Critical Care Medicine, Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado, Aurora, Colorado; and
| | - Susan M Majka
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University, Nashville, Tennesse; Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, Tennessee; Vanderbilt Pulmonary Circulation Center, Vanderbilt University, Nashville, Tennessee; Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennesse;
| |
Collapse
|
50
|
Regulator of G-protein signaling-5 is a marker of hepatic stellate cells and expression mediates response to liver injury. PLoS One 2014; 9:e108505. [PMID: 25290689 PMCID: PMC4188519 DOI: 10.1371/journal.pone.0108505] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 08/22/2014] [Indexed: 12/11/2022] Open
Abstract
Liver fibrosis is mediated by hepatic stellate cells (HSCs), which respond to a variety of cytokine and growth factors to moderate the response to injury and create extracellular matrix at the site of injury. G-protein coupled receptor (GPCR)-mediated signaling, via endothelin-1 (ET-1) and angiotensin II (AngII), increases HSC contraction, migration and fibrogenesis. Regulator of G-protein signaling-5 (RGS5), an inhibitor of vasoactive GPCR agonists, functions to control GPCR-mediated contraction and hypertrophy in pericytes and smooth muscle cells (SMCs). Therefore we hypothesized that RGS5 controls GPCR signaling in activated HSCs in the context of liver injury. In this study, we localize RGS5 to the HSCs and demonstrate that Rgs5 expression is regulated during carbon tetrachloride (CCl4)-induced acute and chronic liver injury in Rgs5LacZ/LacZ reporter mice. Furthermore, CCl4 treated RGS5-null mice develop increased hepatocyte damage and fibrosis in response to CCl4 and have increased expression of markers of HSC activation. Knockdown of Rgs5 enhances ET-1-mediated signaling in HSCs in vitro. Taken together, we demonstrate that RGS5 is a critical regulator of GPCR signaling in HSCs and regulates HSC activation and fibrogenesis in liver injury.
Collapse
|