1
|
Sridhar S, Zhou Y, Ibrahim A, Bertazzo S, Wyss T, Swain A, Maheshwari U, Huang SF, Colonna M, Keller A. Targeting TREM2 signaling shows limited impact on cerebrovascular calcification. Life Sci Alliance 2025; 8:e202402796. [PMID: 39467636 PMCID: PMC11519321 DOI: 10.26508/lsa.202402796] [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: 04/26/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/30/2024] Open
Abstract
Brain calcification, the ectopic mineral deposits of calcium phosphate, is a frequent radiological finding and a diagnostic criterion for primary familial brain calcification. We previously showed that microglia curtail the growth of small vessel calcification via the triggering receptor expressed in myeloid 2 (TREM2) in the Pdgfb ret/ret mouse model of primary familial brain calcification. Because boosting TREM2 function using activating antibodies has been shown to be beneficial in other disease conditions by aiding in microglial clearance of diverse pathologies, we investigated whether administration of a TREM2-activating antibody could mitigate vascular calcification in Pdgfb ret/ret mice. Single-nucleus RNA-sequencing analysis showed that calcification-associated microglia share transcriptional similarities to disease-associated microglia and exhibited activated TREM2 and TGFβ signaling. Administration of a TREM2-activating antibody increased TREM2-dependent microglial deposition of cathepsin K, a collagen-degrading protease, onto calcifications. However, this did not ameliorate the calcification load or alter the mineral composition and the microglial phenotype around calcification. We therefore conclude that targeting microglia with TREM2 agonistic antibodies is insufficient to demineralize and clear vascular calcifications.
Collapse
Affiliation(s)
- Sucheta Sridhar
- https://ror.org/02crff812 Department of Neurosurgery, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- https://ror.org/02crff812 Neuroscience Centre Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Yingyue Zhou
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Adiljan Ibrahim
- https://ror.org/03vagve85 Alector, South San Francisco, CA, USA
| | - Sergio Bertazzo
- https://ror.org/02jx3x895 Department of Medical Physics and Biomedical Engineering, University College London, London, UK
| | - Tania Wyss
- TDS-facility, AGORA Cancer Research Center, Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Amanda Swain
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Upasana Maheshwari
- https://ror.org/02crff812 Department of Neurosurgery, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Sheng-Fu Huang
- https://ror.org/02crff812 Department of Neurosurgery, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Annika Keller
- https://ror.org/02crff812 Department of Neurosurgery, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- https://ror.org/02crff812 Neuroscience Centre Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
2
|
Strell C, Rodríguez-Tomàs E, Östman A. Functional and clinical roles of stromal PDGF receptors in tumor biology. Cancer Metastasis Rev 2024; 43:1593-1609. [PMID: 38980580 DOI: 10.1007/s10555-024-10194-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 06/12/2024] [Indexed: 07/10/2024]
Abstract
PDGF receptors play pivotal roles in both developmental and physiological processes through the regulation of mesenchymal cells involved in paracrine instructive interactions with epithelial or endothelial cells. Tumor biology studies, alongside analyses of patient tissue samples, provide strong indications that the PDGF signaling pathways are also critical in various types of human cancer. This review summarizes experimental findings and correlative studies, which have explored the biological mechanisms and clinical relevance of PDGFRs in mesenchymal cells of the tumor microenvironment. Collectively, these studies support the overall concept that the PDGF system is a critical regulator of tumor growth, metastasis, and drug efficacy, suggesting yet unexploited targeting opportunities. The inter-patient variability in stromal PDGFR expression, as being linked to prognosis and treatment responses, not only indicates the need for stratified approaches in upcoming therapeutic investigations but also implies the potential for the development of PDGFRs as biomarkers of clinical utility, interestingly also in settings outside PDGFR-directed treatments.
Collapse
Affiliation(s)
- Carina Strell
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, Bergen University, Bergen, Norway
| | | | - Arne Östman
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, Bergen University, Bergen, Norway.
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
3
|
Yang L, Han Y, Zhang T, Dong X, Ge J, Roy A, Zhu J, Lu T, Jeya Vandana J, de Silva N, Robertson CC, Xiang JZ, Pan C, Sun Y, Que J, Evans T, Liu C, Wang W, Naji A, Parker SCJ, Schwartz RE, Chen S. Human vascularized macrophage-islet organoids to model immune-mediated pancreatic β cell pyroptosis upon viral infection. Cell Stem Cell 2024; 31:1612-1629.e8. [PMID: 39232561 PMCID: PMC11546835 DOI: 10.1016/j.stem.2024.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 06/05/2024] [Accepted: 08/09/2024] [Indexed: 09/06/2024]
Abstract
There is a paucity of human models to study immune-mediated host damage. Here, we utilized the GeoMx spatial multi-omics platform to analyze immune cell changes in COVID-19 pancreatic autopsy samples, revealing an accumulation of proinflammatory macrophages. Single-cell RNA sequencing (scRNA-seq) analysis of human islets exposed to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or coxsackievirus B4 (CVB4) viruses identified activation of proinflammatory macrophages and β cell pyroptosis. To distinguish viral versus proinflammatory-macrophage-mediated β cell pyroptosis, we developed human pluripotent stem cell (hPSC)-derived vascularized macrophage-islet (VMI) organoids. VMI organoids exhibited enhanced marker expression and function in both β cells and endothelial cells compared with separately cultured cells. Notably, proinflammatory macrophages within VMI organoids induced β cell pyroptosis. Mechanistic investigations highlighted TNFSF12-TNFRSF12A involvement in proinflammatory-macrophage-mediated β cell pyroptosis. This study established hPSC-derived VMI organoids as a valuable tool for studying immune-cell-mediated host damage and uncovered the mechanism of β cell damage during viral exposure.
Collapse
Affiliation(s)
- Liuliu Yang
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; Center for Genomic Health, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Disease, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China; Tianjin Institute of Health Science, Tianjin 301600, China.
| | - Yuling Han
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; Center for Genomic Health, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Tuo Zhang
- Genomic Resource Core Facility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Xue Dong
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Jian Ge
- Columbia Center for Human Development, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Aadita Roy
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Jiajun Zhu
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; Center for Genomic Health, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Tiankun Lu
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; Center for Genomic Health, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - J Jeya Vandana
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; Center for Genomic Health, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Neranjan de Silva
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; Center for Genomic Health, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Catherine C Robertson
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Jenny Z Xiang
- Genomic Resource Core Facility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Chendong Pan
- Genomic Resource Core Facility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Yanjie Sun
- Genomic Resource Core Facility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Jianwen Que
- Columbia Center for Human Development, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Todd Evans
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; Center for Genomic Health, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Chengyang Liu
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Wei Wang
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Ali Naji
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Stephen C J Parker
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA; Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA; Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Robert E Schwartz
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA.
| | - Shuibing Chen
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; Center for Genomic Health, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
4
|
Maniaci A, Briglia M, Allia F, Montalbano G, Romano GL, Zaouali MA, H’mida D, Gagliano C, Malaguarnera R, Lentini M, Graziano ACE, Giurdanella G. The Role of Pericytes in Inner Ear Disorders: A Comprehensive Review. BIOLOGY 2024; 13:802. [PMID: 39452111 PMCID: PMC11504721 DOI: 10.3390/biology13100802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 10/02/2024] [Accepted: 10/06/2024] [Indexed: 10/26/2024]
Abstract
Inner ear disorders, including sensorineural hearing loss, Meniere's disease, and vestibular neuritis, are prevalent conditions that significantly impact the quality of life. Despite their high incidence, the underlying pathophysiology of these disorders remains elusive, and current treatment options are often inadequate. Emerging evidence suggests that pericytes, a type of vascular mural cell specialized to maintain the integrity and function of the microvasculature, may play a crucial role in the development and progression of inner ear disorders. The pericytes are present in the microvasculature of both the cochlea and the vestibular system, where they regulate blood flow, maintain the blood-labyrinth barrier, facilitate angiogenesis, and provide trophic support to neurons. Understanding their role in inner ear disorders may provide valuable insights into the pathophysiology of these conditions and lead to the development of novel diagnostic and therapeutic strategies, improving the standard of living. This comprehensive review aims to provide a detailed overview of the role of pericytes in inner ear disorders, highlighting the anatomy and physiology in the microvasculature, and analyzing the mechanisms that contribute to the development of the disorders. Furthermore, we explore the potential pericyte-targeted therapies, including antioxidant, anti-inflammatory, and angiogenic approaches, as well as gene therapy strategies.
Collapse
Affiliation(s)
- Antonino Maniaci
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
- Department of Surgery, ENT Unit, Asp 7 Ragusa, 97100 Ragusa, Italy
| | - Marilena Briglia
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Fabio Allia
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Giuseppe Montalbano
- Zebrafish Neuromorphology Laboratory, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy;
| | - Giovanni Luca Romano
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Mohamed Amine Zaouali
- Laboratory of Human Genome and Multifactorial Diseases (LR12ES07), Faculty of Pharmacy, University of Monastir, Avicenne Street, 5019 Monastir, Tunisia;
| | - Dorra H’mida
- Department of Cytogenetics and Reproductive Biology, Farhat Hached Hospital, 4021 Sousse, Tunisia;
| | - Caterina Gagliano
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Roberta Malaguarnera
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Mario Lentini
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
- Department of Surgery, ENT Unit, Asp 7 Ragusa, 97100 Ragusa, Italy
| | - Adriana Carol Eleonora Graziano
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Giovanni Giurdanella
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| |
Collapse
|
5
|
Cano E, Schwarzkopf J, Kanda M, Lindberg EL, Hollfinger I, Pogontke C, Braeuning C, Fischer C, Hübner N, Gerhardt H. Intramyocardial Sprouting Tip Cells Specify Coronary Arterialization. Circ Res 2024; 135:671-684. [PMID: 39092506 PMCID: PMC11361357 DOI: 10.1161/circresaha.124.324868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/11/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024]
Abstract
BACKGROUND The elaborate patterning of coronary arteries critically supports the high metabolic activity of the beating heart. How coronary endothelial cells coordinate hierarchical vascular remodeling and achieve arteriovenous specification remains largely unknown. Understanding the molecular and cellular cues that pattern coronary arteries is crucial to develop innovative therapeutic strategies that restore functional perfusion within the ischemic heart. METHODS Single-cell transcriptomics and histological validation were used to delineate heterogeneous transcriptional states of the developing and mature coronary endothelium with a focus on sprouting endothelium and arterial cell specification. Genetic lineage tracing and high-resolution 3-dimensional imaging were used to characterize the origin and mechanisms of coronary angiogenic sprouting, as well as to fate-map selective endothelial lineages. Integration of single-cell transcriptomic data from ischemic adult mouse hearts and human embryonic data served to assess the conservation of transcriptional states across development, disease, and species. RESULTS We discover that coronary arteries originate from cells that have previously transitioned through a specific tip cell phenotype. We identify nonoverlapping intramyocardial and subepicardial tip cell populations with differential gene expression profiles and regulatory pathways. Esm1-lineage tracing confirmed that intramyocardial tip cells selectively contribute to coronary arteries and endocardial tunnels, but not veins. Notably, prearterial cells are detected from development stages to adulthood, increasingly in response to ischemic injury, and in human embryos, suggesting that tip cell-to-artery specification is a conserved mechanism. CONCLUSIONS A tip cell-to-artery specification mechanism drives arterialization of the intramyocardial plexus and endocardial tunnels throughout life and is reactivated upon ischemic injury. Differential sprouting programs govern the formation and specification of the venous and arterial coronary plexus.
Collapse
Affiliation(s)
- Elena Cano
- Integrative Vascular Biology Laboratory (E.C., J.S., I.H., H.G.), Max-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Berlin, Germany (E.C., J.S., N.H., H.G.)
- Charité-Universitätsmedizin, Berlin, Germany (E.C., J.S., N.H., H.G.)
- Department of Animal Biology, University of Málaga, Spain (E.C., C.P.)
- Cardiovascular Development and Disease, Biomedical Research Institute of Malaga and Nanomedicine Platform (IBIMA - BIONAND Platform), Málaga, Spain (E.C., C.P.)
| | - Jennifer Schwarzkopf
- Integrative Vascular Biology Laboratory (E.C., J.S., I.H., H.G.), Max-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Berlin, Germany (E.C., J.S., N.H., H.G.)
- Charité-Universitätsmedizin, Berlin, Germany (E.C., J.S., N.H., H.G.)
| | - Masatoshi Kanda
- Cardiovascular and Metabolic Sciences (M.K., E.L.L., N.H.), Max-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Department of Rheumatology and Clinical Immunology, Sapporo Medical University, Japan (M.K.)
| | - Eric L. Lindberg
- Cardiovascular and Metabolic Sciences (M.K., E.L.L., N.H.), Max-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Department of Medicine, Ludwig-Maximiliams-University Munich, Germany (E.L.L.)
| | - Irene Hollfinger
- Integrative Vascular Biology Laboratory (E.C., J.S., I.H., H.G.), Max-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Cristina Pogontke
- Department of Animal Biology, University of Málaga, Spain (E.C., C.P.)
- Cardiovascular Development and Disease, Biomedical Research Institute of Malaga and Nanomedicine Platform (IBIMA - BIONAND Platform), Málaga, Spain (E.C., C.P.)
| | | | | | - Norbert Hübner
- Cardiovascular and Metabolic Sciences (M.K., E.L.L., N.H.), Max-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Berlin, Germany (E.C., J.S., N.H., H.G.)
- Charité-Universitätsmedizin, Berlin, Germany (E.C., J.S., N.H., H.G.)
| | - Holger Gerhardt
- Integrative Vascular Biology Laboratory (E.C., J.S., I.H., H.G.), Max-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Berlin, Germany (E.C., J.S., N.H., H.G.)
- Charité-Universitätsmedizin, Berlin, Germany (E.C., J.S., N.H., H.G.)
- Berlin Institute of Health (BIH), Germany (H.G.)
| |
Collapse
|
6
|
Yang L, Han Y, Zhang T, Dong X, Ge J, Roy A, Zhu J, Lu T, Vandana JJ, de Silva N, Robertson CC, Xiang JZ, Pan C, Sun Y, Que J, Evans T, Liu C, Wang W, Naji A, Parker SC, Schwartz RE, Chen S. Human Vascularized Macrophage-Islet Organoids to Model Immune-Mediated Pancreatic β cell Pyroptosis upon Viral Infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.05.606734. [PMID: 39149298 PMCID: PMC11326194 DOI: 10.1101/2024.08.05.606734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
There is a paucity of human models to study immune-mediated host damage. Here, we utilized the GeoMx spatial multi-omics platform to analyze immune cell changes in COVID-19 pancreatic autopsy samples, revealing an accumulation of proinflammatory macrophages. Single cell RNA-seq analysis of human islets exposed to SARS-CoV-2 or Coxsackievirus B4 (CVB4) viruses identified activation of proinflammatory macrophages and β cell pyroptosis. To distinguish viral versus proinflammatory macrophage-mediated β cell pyroptosis, we developed human pluripotent stem cell (hPSC)-derived vascularized macrophage-islet (VMI) organoids. VMI organoids exhibited enhanced marker expression and function in both β cells and endothelial cells compared to separately cultured cells. Notably, proinflammatory macrophages within VMI organoids induced β cell pyroptosis. Mechanistic investigations highlighted TNFSF12-TNFRSF12A involvement in proinflammatory macrophage-mediated β cell pyroptosis. This study established hPSC-derived VMI organoids as a valuable tool for studying immune cell-mediated host damage and uncovered mechanism of β cell damage during viral exposure.
Collapse
Affiliation(s)
- Liuliu Yang
- Department of Surgery, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
- Center for Genomic Health, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Disease, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
- Tianjin Institute of Health Science, Tianjin 301600, China
| | - Yuling Han
- Department of Surgery, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
- Center for Genomic Health, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Tuo Zhang
- Genomic Resource Core Facility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Xue Dong
- Department of Surgery, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
| | - Jian Ge
- Columbia Center for Human Development, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Aadita Roy
- Department of Surgery, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
| | - Jiajun Zhu
- Department of Surgery, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
- Center for Genomic Health, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
| | - Tiankun Lu
- Department of Surgery, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
- Center for Genomic Health, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
| | - J. Jeya Vandana
- Department of Surgery, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
- Center for Genomic Health, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
| | - Neranjan de Silva
- Department of Surgery, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
- Center for Genomic Health, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
| | - Catherine C. Robertson
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Jenny Z Xiang
- Genomic Resource Core Facility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Chendong Pan
- Genomic Resource Core Facility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Yanjie Sun
- Genomic Resource Core Facility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Jianwen Que
- Columbia Center for Human Development, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Todd Evans
- Department of Surgery, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
- Center for Genomic Health, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
| | - Chengyang Liu
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Wei Wang
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Ali Naji
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Stephen C.J. Parker
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Robert E. Schwartz
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA. New York 10021, USA
| | - Shuibing Chen
- Department of Surgery, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
- Center for Genomic Health, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
| |
Collapse
|
7
|
Perez-Gutierrez L, Li P, Ferrara N. Endothelial cell diversity: the many facets of the crystal. FEBS J 2024; 291:3287-3302. [PMID: 36266750 DOI: 10.1111/febs.16660] [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: 07/17/2022] [Revised: 10/03/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022]
Abstract
Endothelial cells (ECs) form the inner lining of blood vessels and play crucial roles in angiogenesis. While it has been known for a long time that there are considerable differences among ECs from lymphatic and blood vessels, as well as among arteries, veins and capillaries, the full repertoire of endothelial diversity is only beginning to be elucidated. It has become apparent that the role of ECs is not just limited to their exchange functions. Indeed, a multitude of organ-specific functions, including release of growth factors, regulation of immune functions, have been linked to ECs. Recent years have seen a surge into the identification of spatiotemporal molecular and functional heterogeneity of ECs, supported by technologies such as single-cell RNA sequencing (scRNA-seq), lineage tracing and intersectional genetics. Together, these techniques have spurred the generation of epigenomic, transcriptomic and proteomic signatures of ECs. It is now clear that ECs across organs and in different vascular beds, but even within the same vessel, have unique molecular identities and employ specialized molecular mechanisms to fulfil highly specialized needs. Here, we focus on the molecular heterogeneity of the endothelium in different organs and pathological conditions.
Collapse
Affiliation(s)
- Lorena Perez-Gutierrez
- Department of Pathology, Moores Cancer Center, University of California, San Diego, CA, USA
| | - Pin Li
- Department of Pathology, Moores Cancer Center, University of California, San Diego, CA, USA
| | - Napoleone Ferrara
- Department of Pathology, Moores Cancer Center, University of California, San Diego, CA, USA
| |
Collapse
|
8
|
Huang W, Zhou H, He Y, Wang A, Wang B, Chen Y, Liu C, Wang H, Xie W, Kong H. A novel PDGFR inhibitor WQ-C-401 prevents pulmonary vascular remodeling in rats with monocrotaline-induced pulmonary arterial hypertension. Exp Cell Res 2024; 441:114154. [PMID: 38996959 DOI: 10.1016/j.yexcr.2024.114154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 07/07/2024] [Accepted: 07/09/2024] [Indexed: 07/14/2024]
Abstract
Platelet-derived growth factor (PDGF) is one of the most important cytokines associated with pulmonary vascular remodeling in pulmonary arterial hypertension (PAH). PDGF receptor (PDGFR) inhibition exerted therapeutic effects on PAH in clinical trials, but serious side effects warrant the withdrawal of existing drugs. In this study, a novel highly selective PDGFR inhibitor WQ-C-401 was developed, and its effects on PDGFR signaling pathway and pulmonary vascular remodeling in PAH were investigated. Cell proliferation assays and Western blot analysis of PDGFRα/β phosphorylation showed that WQ-C-401 inhibited PDGFR-mediated cell proliferation assay and suppressed PDGFR phosphorylation in a concentration-dependent manner. DiscoverX's KinomeScanTM technology confirmed the good kinome selectivity of WQ-C-401 (S score (1) of PDGFR = (0.01)). In monocrotaline (MCT)-induced PAH rats, intragastric administration of WQ-C-401 (25, 50, 100 mg/kg/d) or imatinib (50 mg/kg/d, positive control) significantly decreased right ventricular systolic pressure (RVSP). Histological analysis demonstrated that WQ-C-401 inhibited pulmonary vascular remodeling by reducing muscularization and fibrosis, as well as alleviated right ventricular hypertrophy in MCT-treated rats. In addition, WQ-C-401 suppressed MCT-induced cell hyperproliferation and CD68+ macrophage infiltration around the pulmonary artery. In vitro, WQ-C-401 inhibited PDGF-BB-induced proliferation and migration of human pulmonary arterial smooth muscle cells (PASMCs). Moreover, Western blot analysis showed that WQ-C-401 concertration-dependently inhibited PDGF-BB-induced phosphorylation of ERK1/2 and PDGFRβ Y751, decreased collagen Ⅰ synthesis and increased alpha smooth muscle actin (α-SMA) expression in PASMCs. Collectively, our results suggest that WQ-C-401 is a selective and potent PDGFR inhibitor which could be a promising drug for the therapeutics of PAH by preventing pulmonary vascular remodeling.
Collapse
MESH Headings
- Animals
- Monocrotaline
- Vascular Remodeling/drug effects
- Rats
- Cell Proliferation/drug effects
- Male
- Rats, Sprague-Dawley
- Pulmonary Arterial Hypertension/drug therapy
- Pulmonary Arterial Hypertension/chemically induced
- Pulmonary Arterial Hypertension/metabolism
- Pulmonary Arterial Hypertension/pathology
- Humans
- Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors
- Receptors, Platelet-Derived Growth Factor/metabolism
- Phosphorylation/drug effects
- Pulmonary Artery/drug effects
- Pulmonary Artery/pathology
- Pulmonary Artery/metabolism
- Signal Transduction/drug effects
- Hypertension, Pulmonary/chemically induced
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/prevention & control
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/metabolism
- Protein Kinase Inhibitors/pharmacology
- Receptor, Platelet-Derived Growth Factor beta/metabolism
- Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors
Collapse
Affiliation(s)
- Wen Huang
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, Jiangsu, PR China
| | - Hong Zhou
- Department of Pulmonary & Critical Care Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi 214023, Jiangsu, PR China
| | - Yiting He
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, Jiangsu, PR China
| | - Aoli Wang
- Anhui Province Key Laboratory of Medical Physics & Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, Anhui, PR China; Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, Anhui, PR China
| | - Beilei Wang
- Anhui Province Key Laboratory of Medical Physics & Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, Anhui, PR China; Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, Anhui, PR China
| | - Yongfei Chen
- Anhui Province Key Laboratory of Medical Physics & Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, Anhui, PR China; Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, Anhui, PR China
| | - Chenyang Liu
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, Jiangsu, PR China
| | - Hong Wang
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, Jiangsu, PR China
| | - Weiping Xie
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, Jiangsu, PR China.
| | - Hui Kong
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, Jiangsu, PR China.
| |
Collapse
|
9
|
Sheng X, Zhang C, Zhao J, Xu J, Zhang P, Ding Q, Zhang J. Microvascular destabilization and intricated network of the cytokines in diabetic retinopathy: from the perspective of cellular and molecular components. Cell Biosci 2024; 14:85. [PMID: 38937783 PMCID: PMC11212265 DOI: 10.1186/s13578-024-01269-7] [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: 04/18/2024] [Accepted: 06/19/2024] [Indexed: 06/29/2024] Open
Abstract
Microvascular destabilization is the primary cause of the inner blood-retinal barrier (iBRB) breakdown and increased vascular leakage in diabetic retinopathy (DR). Microvascular destabilization results from the combinational effects of increased levels of growth factors and cytokines, involvement of inflammation, and the changed cell-to-cell interactions, especially the loss of endothelial cells and pericytes, due to hyperglycemia and hypoxia. As the manifestation of microvascular destabilization, the fluid transports via paracellular and transcellular routes increase due to the disruption of endothelial intercellular junctional complexes and/or the altered caveolar transcellular transport across the retinal vascular endothelium. With diabetes progression, the functional and the structural changes of the iBRB components, including the cellular and noncellular components, further facilitate and aggravate microvascular destabilization, resulting in macular edema, the neuroretinal damage and the dysfunction of retinal inner neurovascular unit (iNVU). Although there have been considerable recent advances towards a better understanding of the complex cellular and molecular network underlying the microvascular destabilization, some still remain to be fully elucidated. Recent data indicate that targeting the intricate signaling pathways may allow to against the microvascular destabilization. Therefore, efforts have been made to better clarify the cellular and molecular mechanisms that are involved in the microvascular destabilization in DR. In this review, we discuss: (1) the brief introduction of DR and microvascular destabilization; (2) the cellular and molecular components of iBRB and iNVU, and the breakdown of iBRB; (3) the matrix and cell-to-cell contacts to maintain microvascular stabilization, including the endothelial glycocalyx, basement membrane, and various cell-cell interactions; (4) the molecular mechanisms mediated cell-cell contacts and vascular cell death; (5) the altered cytokines and signaling pathways as well as the intricate network of the cytokines involved in microvascular destabilization. This comprehensive review aimed to provide the insights for microvascular destabilization by targeting the key molecules or specific iBRB cells, thus restoring the function and structure of iBRB and iNVU, to treat DR.
Collapse
Affiliation(s)
- Xia Sheng
- People's Hospital of Huangdao District, Qingdao, Shandong Province, China
| | - Chunmei Zhang
- People's Hospital of Huangdao District, Qingdao, Shandong Province, China
| | - Jiwei Zhao
- People's Hospital of Huangdao District, Qingdao, Shandong Province, China
| | - Jianping Xu
- People's Hospital of Huangdao District, Qingdao, Shandong Province, China.
| | - Peng Zhang
- People's Hospital of Huangdao District, Qingdao, Shandong Province, China.
| | - Quanju Ding
- People's Hospital of Huangdao District, Qingdao, Shandong Province, China.
| | - Jingfa Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, National Clinical Research Center for Eye Diseases, Shanghai, China.
- The International Eye Research Institute of The Chinese University of Hong Kong (Shenzhen), Shenzhen, China.
- C-MER (Shenzhen) Dennis Lam Eye Hospital, Shenzhen, China.
- C-MER International Eye Care Group, C-MER Dennis Lam & Partners Eye Center, Hong Kong, China.
| |
Collapse
|
10
|
Amoedo-Leite C, Parv K, Testini C, Herrera-Hidalgo C, Xu F, Giraud A, Malaquias M, Fasterius E, Holl D, Seignez C, Göritz C, Christoffersson G, Phillipson M. Macrophages upregulate mural cell-like markers and support healing of ischemic injury by adopting functions important for vascular support. NATURE CARDIOVASCULAR RESEARCH 2024; 3:685-700. [PMID: 39196227 PMCID: PMC11358018 DOI: 10.1038/s44161-024-00478-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 04/25/2024] [Indexed: 08/29/2024]
Abstract
Sterile inflammation after injury is important for tissue restoration. In injured human and mouse tissues, macrophages were recently found to accumulate perivascularly. This study investigates if macrophages adopt a mural cell phenotype important for restoration after ischemic injury. Single-cell RNA sequencing of fate-mapped macrophages from ischemic mouse muscles demonstrates a macrophage-toward-mural cell switch of a subpopulation of macrophages with downregulated myeloid cell genes and upregulated mural cell genes, including PDGFRβ. This observation was further strengthened when including unspliced transcripts in the analysis. The macrophage switch was proven functionally relevant, as induction of macrophage-specific PDGFRβ deficiency prevented their perivascular macrophage phenotype, impaired vessel maturation and increased vessel leakiness, which ultimately reduced limb function. In conclusion, macrophages in adult ischemic tissue were demonstrated to undergo a cellular program to morphologically, transcriptomically and functionally resemble mural cells while weakening their macrophage identity. The macrophage-to-mural cell-like phenotypic switch is crucial for restoring tissue function and warrants further exploration as a potential target for immunotherapies to enhance healing.
Collapse
Affiliation(s)
| | - Kristel Parv
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Chiara Testini
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | | | - Feifei Xu
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Antoine Giraud
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Marta Malaquias
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Erik Fasterius
- National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Stockholm University, Solna, Sweden
| | - Daniel Holl
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Cedric Seignez
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Christian Göritz
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, Hong Kong
| | - Gustaf Christoffersson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Mia Phillipson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden.
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
11
|
Lin Y, Gahn J, Banerjee K, Dobreva G, Singhal M, Dubrac A, Ola R. Role of endothelial PDGFB in arterio-venous malformations pathogenesis. Angiogenesis 2024; 27:193-209. [PMID: 38070064 PMCID: PMC11021264 DOI: 10.1007/s10456-023-09900-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 11/05/2023] [Indexed: 04/17/2024]
Abstract
Arterial-venous malformations (AVMs) are direct connections between arteries and veins without an intervening capillary bed. Either familial inherited or sporadically occurring, localized pericytes (PCs) drop is among the AVMs' hallmarks. Whether impaired PC coverage triggers AVMs or it is a secondary event is unclear. Here we evaluated the role of the master regulator of PC recruitment, Platelet derived growth factor B (PDGFB) in AVM pathogenesis. Using tamoxifen-inducible deletion of Pdgfb in endothelial cells (ECs), we show that disruption of EC Pdgfb-mediated PC recruitment and maintenance leads to capillary enlargement and organotypic AVM-like structures. These vascular lesions contain non-proliferative hyperplastic, hypertrophic and miss-oriented capillary ECs with an altered capillary EC fate identity. Mechanistically, we propose that PDGFB maintains capillary EC size and caliber to limit hemodynamic changes, thus restricting expression of Krüppel like factor 4 and activation of Bone morphogenic protein, Transforming growth factor β and NOTCH signaling in ECs. Furthermore, our study emphasizes that inducing or activating PDGFB signaling may be a viable therapeutic approach for treating vascular malformations.
Collapse
Affiliation(s)
- Yanzhu Lin
- Experimental Pharmacology Mannheim (EPM), European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Johannes Gahn
- Experimental Pharmacology Mannheim (EPM), European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Kuheli Banerjee
- Experimental Pharmacology Mannheim (EPM), European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Gergana Dobreva
- Department of Cardiovascular Genomics and Epigenomics, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- German Centre for Cardiovascular Research (DZHK), Heidelberg, Germany
| | - Mahak Singhal
- Laboratory of AngioRhythms, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Alexandre Dubrac
- Centre de Recherche, CHU St. Justine, Montreal, QC, H3T 1C5, Canada
- Département de Pathologie et Biologie Cellulaire, Université de Montréal, Montreal, QC, H3T 1J4, Canada
| | - Roxana Ola
- Experimental Pharmacology Mannheim (EPM), European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
12
|
Liu G, Shu W, Chen Y, Fu Y, Fang S, Zheng H, Cheng W, Lin Q, Hu Y, Jiang N, Yu B. Bone-derived PDGF-BB enhances hippocampal non-specific transcytosis through microglia-endothelial crosstalk in HFD-induced metabolic syndrome. J Neuroinflammation 2024; 21:111. [PMID: 38685040 PMCID: PMC11057146 DOI: 10.1186/s12974-024-03097-5] [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: 08/28/2023] [Accepted: 04/10/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND It is well known that high-fat diet (HFD)-induced metabolic syndrome plays a crucial role in cognitive decline and brain-blood barrier (BBB) breakdown. However, whether the bone-brain axis participates in this pathological process remains unknown. Here, we report that platelet-derived growth factor-BB (PDGF-BB) secretion by preosteoclasts in the bone accelerates neuroinflammation. The expression of alkaline phosphatase (ALPL), a nonspecific transcytosis marker, was upregulated during HFD challenge. MAIN BODY Preosteoclast-specific Pdgfb transgenic mice with high PDGF-BB concentrations in the circulation recapitulated the HFD-induced neuroinflammation and transcytosis shift. Preosteoclast-specific Pdgfb knockout mice were partially rescued from hippocampal neuroinflammation and transcytosis shifts in HFD-challenged mice. HFD-induced PDGF-BB elevation aggravated microglia-associated neuroinflammation and interleukin-1β (IL-1β) secretion, which increased ALPL expression and transcytosis shift through enhancing protein 1 (SP1) translocation in endothelial cells. CONCLUSION Our findings confirm the role of bone-secreted PDGF-BB in neuroinflammation and the transcytosis shift in the hippocampal region during HFD challenge and identify a novel mechanism of microglia-endothelial crosstalk in HFD-induced metabolic syndrome.
Collapse
Affiliation(s)
- Guanqiao Liu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wen Shu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Trauma Orthopedics, Liuzhou People's Hospital, Liuzhou, China
| | - Yingqi Chen
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yong Fu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Trauma Center, Department of Orthopaedic Trauma, The Second Affiliated Hospital of Hengyang Medical College, South China University, Hengyang, China
| | - Shuai Fang
- Trauma Center, Department of Orthopaedic Trauma, The Second Affiliated Hospital of Hengyang Medical College, South China University, Hengyang, China
| | - Haonan Zheng
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weike Cheng
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qingrong Lin
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yanjun Hu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Nan Jiang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Bin Yu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
13
|
Hirobe T. Role of Dermal Factors Involved in Regulating the Melanin and Melanogenesis of Mammalian Melanocytes in Normal and Abnormal Skin. Int J Mol Sci 2024; 25:4560. [PMID: 38674144 PMCID: PMC11049857 DOI: 10.3390/ijms25084560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/13/2024] [Accepted: 04/14/2024] [Indexed: 04/28/2024] Open
Abstract
Mammalian melanin is produced in melanocytes and accumulated in melanosomes. Melanogenesis is supported by many factors derived from the surrounding tissue environment, such as the epidermis, dermis, and subcutaneous tissue, in addition to numerous melanogenesis-related genes. The roles of these genes have been fully investigated and the molecular analysis has been performed. Moreover, the role of paracrine factors derived from epidermis has also been studied. However, the role of dermis has not been fully studied. Thus, in this review, dermis-derived factors including soluble and insoluble components were overviewed and discussed in normal and abnormal circumstances. Dermal factors play an important role in the regulation of melanogenesis in the normal and abnormal mammalian skin.
Collapse
Affiliation(s)
- Tomohisa Hirobe
- Department of Molecular Imaging and Theranostics, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| |
Collapse
|
14
|
Lin PK, Sun Z, Davis GE. Defining the Functional Influence of Endothelial Cell-Expressed Oncogenic Activating Mutations on Vascular Morphogenesis and Capillary Assembly. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:574-598. [PMID: 37838010 PMCID: PMC10988768 DOI: 10.1016/j.ajpath.2023.08.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/02/2023] [Accepted: 08/15/2023] [Indexed: 10/16/2023]
Abstract
This study sought to define key molecules and signals controlling major steps in vascular morphogenesis, and how these signals regulate pericyte recruitment and pericyte-induced basement membrane deposition. The morphogenic impact of endothelial cell (EC) expression of activating mutants of Kirsten rat sarcoma virus (kRas), mitogen-activated protein kinase 1 (Mek1), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), Akt serine/threonine kinase 1 (Akt1), Ras homolog enriched in brain (Rheb) Janus kinase 2 (Jak2), or signal transducer and activator of transcription 3 (Stat3) expression versus controls was evaluated, along with EC signaling events, pharmacologic inhibitor assays, and siRNA suppression experiments. Primary stimulators of EC lumen formation included kRas, Akt1, and Mek1, whereas PIK3CA and Akt1 stimulated a specialized type of cystic lumen formation. In contrast, the key drivers of EC sprouting behavior were Jak2, Stat3, Mek1, PIK3CA, and mammalian target of rapamycin (mTor). These conclusions are further supported by pharmacologic inhibitor and siRNA suppression experiments. EC expression of active Akt1, kRas, and PIK3CA led to markedly dysregulated lumen formation coupled to strongly inhibited pericyte recruitment and basement membrane deposition. For example, activated Akt1 expression in ECs excessively stimulated lumen formation, decreased EC sprouting behavior, and showed minimal pericyte recruitment with reduced mRNA expression of platelet-derived growth factor-BB, platelet-derived growth factor-DD, and endothelin-1, critical EC-derived factors known to stimulate pericyte invasion. The study identified key signals controlling fundamental steps in capillary morphogenesis and maturation and provided mechanistic details on why EC activating mutations induced a capillary deficiency state with abnormal lumens, impaired pericyte recruitment, and basement deposition: predisposing stimuli for the development of vascular malformations.
Collapse
Affiliation(s)
- Prisca K Lin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - Zheying Sun
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - George E Davis
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida.
| |
Collapse
|
15
|
Shrouder JJ, Calandra GM, Filser S, Varga DP, Besson-Girard S, Mamrak U, Dorok M, Bulut-Impraim B, Seker FB, Gesierich B, Laredo F, Wehn AC, Khalin I, Bayer P, Liesz A, Gokce O, Plesnila N. Continued dysfunction of capillary pericytes promotes no-reflow after experimental stroke in vivo. Brain 2024; 147:1057-1074. [PMID: 38153327 DOI: 10.1093/brain/awad401] [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: 03/17/2023] [Revised: 11/15/2023] [Accepted: 11/16/2023] [Indexed: 12/29/2023] Open
Abstract
Incomplete reperfusion of the microvasculature ('no-reflow') after ischaemic stroke damages salvageable brain tissue. Previous ex vivo studies suggest pericytes are vulnerable to ischaemia and may exacerbate no-reflow, but the viability of pericytes and their association with no-reflow remains under-explored in vivo. Using longitudinal in vivo two-photon single-cell imaging over 7 days, we showed that 87% of pericytes constrict during cerebral ischaemia and remain constricted post reperfusion, and 50% of the pericyte population are acutely damaged. Moreover, we revealed ischaemic pericytes to be fundamentally implicated in capillary no-reflow by limiting and arresting blood flow within the first 24 h post stroke. Despite sustaining acute membrane damage, we observed that over half of all cortical pericytes survived ischaemia and responded to vasoactive stimuli, upregulated unique transcriptomic profiles and replicated. Finally, we demonstrated the delayed recovery of capillary diameter by ischaemic pericytes after reperfusion predicted vessel reconstriction in the subacute phase of stroke. Cumulatively, these findings demonstrate that surviving cortical pericytes remain both viable and promising therapeutic targets to counteract no-reflow after ischaemic stroke.
Collapse
Affiliation(s)
- Joshua James Shrouder
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Gian Marco Calandra
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, 81377 Munich, Germany
| | - Severin Filser
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- Core Research Facilities and Services-Light Microscope Facility, German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Daniel Peter Varga
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Simon Besson-Girard
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Uta Mamrak
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, 81377 Munich, Germany
| | - Maximilian Dorok
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, 81377 Munich, Germany
| | - Buket Bulut-Impraim
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Fatma Burcu Seker
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Benno Gesierich
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Fabio Laredo
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, 81377 Munich, Germany
| | - Antonia Clarissa Wehn
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- Department of Neurosurgery, LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, 81377 Munich, Germany
| | - Igor Khalin
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- Normandie University, UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institute Blood and Brain @ Caen-Normandie (BB@C), 14000 Caen, France
| | - Patrick Bayer
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, 81377 Munich, Germany
| | - Arthur Liesz
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Ozgun Gokce
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| |
Collapse
|
16
|
Lund H, Hunt MA, Kurtović Z, Sandor K, Kägy PB, Fereydouni N, Julien A, Göritz C, Vazquez-Liebanas E, Andaloussi Mäe M, Jurczak A, Han J, Zhu K, Harris RA, Lampa J, Graversen JH, Etzerodt A, Haglund L, Yaksh TL, Svensson CI. CD163+ macrophages monitor enhanced permeability at the blood-dorsal root ganglion barrier. J Exp Med 2024; 221:e20230675. [PMID: 38117255 PMCID: PMC10733632 DOI: 10.1084/jem.20230675] [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: 04/20/2023] [Revised: 10/04/2023] [Accepted: 11/27/2023] [Indexed: 12/21/2023] Open
Abstract
In dorsal root ganglia (DRG), macrophages reside close to sensory neurons and have largely been explored in the context of pain, nerve injury, and repair. However, we discovered that most DRG macrophages interact with and monitor the vasculature by sampling macromolecules from the blood. Characterization of the DRG vasculature revealed a specialized endothelial bed that transformed in molecular, structural, and permeability properties along the arteriovenous axis and was covered by macrophage-interacting pericytes and fibroblasts. Macrophage phagocytosis spatially aligned with peak endothelial permeability, a process regulated by enhanced caveolar transcytosis in endothelial cells. Profiling the DRG immune landscape revealed two subsets of perivascular macrophages with distinct transcriptome, turnover, and function. CD163+ macrophages self-maintained locally, specifically participated in vasculature monitoring, displayed distinct responses during peripheral inflammation, and were conserved in mouse and man. Our work provides a molecular explanation for the permeability of the blood-DRG barrier and identifies an unappreciated role of macrophages as integral components of the DRG-neurovascular unit.
Collapse
Affiliation(s)
- Harald Lund
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Matthew A. Hunt
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Zerina Kurtović
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Kancera AB, Karolinska Institutet Science Park, Stockholm, Sweden
| | - Katalin Sandor
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Paul B. Kägy
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Noah Fereydouni
- Department of Medicine, Rheumatology Unit, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Anais Julien
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Christian Göritz
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Elisa Vazquez-Liebanas
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Maarja Andaloussi Mäe
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Alexandra Jurczak
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jinming Han
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Keying Zhu
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Robert A. Harris
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Jon Lampa
- Department of Medicine, Rheumatology Unit, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | | | - Anders Etzerodt
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Lisbet Haglund
- Division of Orthopaedic Surgery, Department of Surgery, McGill University, Montreal, Canada
| | - Tony L. Yaksh
- Department of Anesthesiology, University of California, San Diego, CA, USA
| | - Camilla I. Svensson
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
17
|
Simmonds SJ, Grootaert MOJ, Cuijpers I, Carai P, Geuens N, Herwig M, Baatsen P, Hamdani N, Luttun A, Heymans S, Jones EAV. Pericyte loss initiates microvascular dysfunction in the development of diastolic dysfunction. EUROPEAN HEART JOURNAL OPEN 2024; 4:oead129. [PMID: 38174347 PMCID: PMC10763525 DOI: 10.1093/ehjopen/oead129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 11/27/2023] [Accepted: 11/30/2023] [Indexed: 01/05/2024]
Abstract
Aims Microvascular dysfunction has been proposed to drive heart failure with preserved ejection fraction (HFpEF), but the initiating molecular and cellular events are largely unknown. Our objective was to determine when microvascular alterations in HFpEF begin, how they contribute to disease progression, and how pericyte dysfunction plays a role herein. Methods and results Microvascular dysfunction, characterized by inflammatory activation, loss of junctional barrier function, and altered pericyte-endothelial crosstalk, was assessed with respect to the development of cardiac dysfunction, in the Zucker fatty and spontaneously hypertensive (ZSF1) obese rat model of HFpEF at three time points: 6, 14, and 21 weeks of age. Pericyte loss was the earliest and strongest microvascular change, occurring before prominent echocardiographic signs of diastolic dysfunction were present. Pericytes were shown to be less proliferative and had a disrupted morphology at 14 weeks in the obese ZSF1 animals, who also exhibited an increased capillary luminal diameter and disrupted endothelial junctions. Microvascular dysfunction was also studied in a mouse model of chronic reduction in capillary pericyte coverage (PDGF-Bret/ret), which spontaneously developed many aspects of diastolic dysfunction. Pericytes exposed to oxidative stress in vitro showed downregulation of cell cycle-associated pathways and induced a pro-inflammatory state in endothelial cells upon co-culture. Conclusion We propose pericytes are important for maintaining endothelial cell function, where loss of pericytes enhances the reactivity of endothelial cells to inflammatory signals and promotes microvascular dysfunction, thereby accelerating the development of HFpEF.
Collapse
Affiliation(s)
- Steven J Simmonds
- Centre for Molecular and Vascular Biology, KU Leuven, Herestraat 49, bus 911, Leuven 3000, Belgium
| | - Mandy O J Grootaert
- Centre for Molecular and Vascular Biology, KU Leuven, Herestraat 49, bus 911, Leuven 3000, Belgium
| | - Ilona Cuijpers
- Centre for Molecular and Vascular Biology, KU Leuven, Herestraat 49, bus 911, Leuven 3000, Belgium
- Department of Cardiology, Maastricht University, CARIM School for Cardiovascular Diseases, Universiteitssingel 50, Maastricht 6229 ER, The Netherlands
| | - Paolo Carai
- Centre for Molecular and Vascular Biology, KU Leuven, Herestraat 49, bus 911, Leuven 3000, Belgium
| | - Nadeche Geuens
- Centre for Molecular and Vascular Biology, KU Leuven, Herestraat 49, bus 911, Leuven 3000, Belgium
| | - Melissa Herwig
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum 44801, Germany
- Molecular and Experimental Cardiology, Institut für Forschung und Lehre (IFL), Ruhr University Bochum, Bochum, Germany
- Department of Cardiology, St.Josef-Hospital, Ruhr University Bochum, Bochum, Germany
| | - Pieter Baatsen
- VIB-KU Leuven, Center for Brain and Disease Research, Electron Microscopy Platform & VIB Bioimaging Core, Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Nazha Hamdani
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum 44801, Germany
- Molecular and Experimental Cardiology, Institut für Forschung und Lehre (IFL), Ruhr University Bochum, Bochum, Germany
- Department of Cardiology, St.Josef-Hospital, Ruhr University Bochum, Bochum, Germany
| | - Aernout Luttun
- Centre for Molecular and Vascular Biology, KU Leuven, Herestraat 49, bus 911, Leuven 3000, Belgium
| | - Stephane Heymans
- Centre for Molecular and Vascular Biology, KU Leuven, Herestraat 49, bus 911, Leuven 3000, Belgium
- Department of Cardiology, Maastricht University, CARIM School for Cardiovascular Diseases, Universiteitssingel 50, Maastricht 6229 ER, The Netherlands
| | - Elizabeth A V Jones
- Centre for Molecular and Vascular Biology, KU Leuven, Herestraat 49, bus 911, Leuven 3000, Belgium
- Department of Cardiology, Maastricht University, CARIM School for Cardiovascular Diseases, Universiteitssingel 50, Maastricht 6229 ER, The Netherlands
| |
Collapse
|
18
|
An X, Ma X, Liu H, Song J, Wei T, Zhang R, Zhan X, Li H, Zhou J. Inhibition of PDGFRβ alleviates endothelial cell apoptotic injury caused by DRP-1 overexpression and mitochondria fusion failure after mitophagy. Cell Death Dis 2023; 14:756. [PMID: 37980402 PMCID: PMC10657461 DOI: 10.1038/s41419-023-06272-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 10/23/2023] [Accepted: 11/03/2023] [Indexed: 11/20/2023]
Abstract
Kawasaki disease (KD), described as "mucocutaneous lymph node syndrome", affects infants and toddlers. Patients with KD suffer from an inflammatory cascade leading to vasculitis with a predilection for coronary arteries. While the symptoms and pathogenesis of KD have received more and more attention, the precise mechanisms are still debated. Researches show that endothelial dysfunction process in KD leads to arterial damage and affect clinical outcome. In this study, we constructed a Candida albicans water soluble fraction (CAWS)-induced KD murine model and penetrated investigating the mechanisms behind endothelial dysfunction. CAWS-induced mice presented remarkably elevated vascular endothelial cell growth factor (VEGF) levels. Abundant expression of VEGF was documented in all vessels that showed edema from acute KD. It has been reported that Platelet-derived growth factor (PDGF) co-expression normalizes VEGF-induced aberrant angiogenesis. Hyperexpression of PDGFRβ was induced in the thickened medial layer and vascular endothelium of KD mice. Masitinib (Mas) is an oral tyrosine kinase inhibitor of numerous targets, which can selectively target PDGFR signaling. We set out to explore whether Mas could regulate coronary pathology in KD. Mas administration significantly reduced the VEGF-induced endothelial cells migration. NOX4 was activated in vascular endothelial cells to produce more ROS. Mitochondrial dysregulated fission and mitophagy caused by DRP-1 overexpression precipitated the arterial endothelial cells injury. Here, mitophagy seemed to work as the driving force of DRP-1/Bak/BNIP3-dependent endothelial cells apoptosis. In summary, how mitophagy is regulated by DRP-1 under pathologic status is critical and complex, which may contribute to the development of specific therapeutic interventions in cardiovascular diseases patients, for example Masatinib, the inhibitor of PDGFRβ. FACTS AND QUESTIONS: Kawasaki disease causing systemic vasculitis, affects infants and toddlers. Coronary artery injury remains the major causes of morbidity and mortality. DRP-1 overexpression induces DRP-1/Bak/BNIP3-dependent endothelial cells apoptosis. PDGFRβ was high-expressed in the thickened medial layer of CAWS-induced KD mice. Inhibition of PDGFRβ signaling alleviates arterial endothelial cells injury.
Collapse
Affiliation(s)
- Xiaohong An
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
- Yunnan Characteristic Plant Extraction Laboratory, Yunnan Yunke Characteristic Plant Extraction Laboratory Co., Ltd, Kunming, 650106, China
| | - Xiao Ma
- Yunnan Characteristic Plant Extraction Laboratory, Yunnan Yunke Characteristic Plant Extraction Laboratory Co., Ltd, Kunming, 650106, China
| | - Heng Liu
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, 671000, China
| | - Jing Song
- Laboratory Animal Center, Xiamen University, Xiamen, 361102, China
| | - Tiange Wei
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Rongzhan Zhang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiao Zhan
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Hongyang Li
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China.
| | - Jia Zhou
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
19
|
Alarcon-Martinez L, Shiga Y, Villafranca-Baughman D, Cueva Vargas JL, Vidal Paredes IA, Quintero H, Fortune B, Danesh-Meyer H, Di Polo A. Neurovascular dysfunction in glaucoma. Prog Retin Eye Res 2023; 97:101217. [PMID: 37778617 DOI: 10.1016/j.preteyeres.2023.101217] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/23/2023] [Accepted: 09/25/2023] [Indexed: 10/03/2023]
Abstract
Retinal ganglion cells, the neurons that die in glaucoma, are endowed with a high metabolism requiring optimal provision of oxygen and nutrients to sustain their activity. The timely regulation of blood flow is, therefore, essential to supply firing neurons in active areas with the oxygen and glucose they need for energy. Many glaucoma patients suffer from vascular deficits including reduced blood flow, impaired autoregulation, neurovascular coupling dysfunction, and blood-retina/brain-barrier breakdown. These processes are tightly regulated by a community of cells known as the neurovascular unit comprising neurons, endothelial cells, pericytes, Müller cells, astrocytes, and microglia. In this review, the neurovascular unit takes center stage as we examine the ability of its members to regulate neurovascular interactions and how their function might be altered during glaucomatous stress. Pericytes receive special attention based on recent data demonstrating their key role in the regulation of neurovascular coupling in physiological and pathological conditions. Of particular interest is the discovery and characterization of tunneling nanotubes, thin actin-based conduits that connect distal pericytes, which play essential roles in the complex spatial and temporal distribution of blood within the retinal capillary network. We discuss cellular and molecular mechanisms of neurovascular interactions and their pathophysiological implications, while highlighting opportunities to develop strategies for vascular protection and regeneration to improve functional outcomes in glaucoma.
Collapse
Affiliation(s)
- Luis Alarcon-Martinez
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada; Centre for Eye Research Australia, University of Melbourne, Melbourne, Australia
| | - Yukihiro Shiga
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada
| | - Deborah Villafranca-Baughman
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada
| | - Jorge L Cueva Vargas
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada
| | - Isaac A Vidal Paredes
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada
| | - Heberto Quintero
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada
| | - Brad Fortune
- Discoveries in Sight Research Laboratories, Devers Eye Institute and Legacy Research Institute, Legacy Healthy, Portland, OR, USA
| | - Helen Danesh-Meyer
- Department of Ophthalmology, New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Adriana Di Polo
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada.
| |
Collapse
|
20
|
Del Gaudio F, Liu D, Andaloussi Mäe M, Braune EB, Hansson EM, Wang QD, Betsholtz C, Lendahl U. Left ventricular hypertrophy and metabolic resetting in the Notch3-deficient adult mouse heart. Sci Rep 2023; 13:15022. [PMID: 37699967 PMCID: PMC10497627 DOI: 10.1038/s41598-023-42010-7] [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: 07/18/2023] [Accepted: 09/04/2023] [Indexed: 09/14/2023] Open
Abstract
The heart depends on a functional vasculature for oxygenation and transport of nutrients, and it is of interest to learn how primary impairment of the vasculature can indirectly affect cardiac function and heart morphology. Notch3-deficiency causes vascular smooth muscle cell (VSMC) loss in the vasculature but the consequences for the heart remain largely elusive. Here, we demonstrate that Notch3-/- mice have enlarged hearts with left ventricular hypertrophy and mild fibrosis. Cardiomyocytes were hypertrophic but not hyperproliferative, and the expression of several cardiomyocyte markers, including Tnt2, Myh6, Myh7 and Actn2, was altered. Furthermore, expression of genes regulating the metabolic status of the heart was affected: both Pdk4 and Cd36 were downregulated, indicating a metabolic switch from fatty acid oxidation to glucose consumption. Notch3-/- mice furthermore showed lower liver lipid content. Notch3 was expressed in heart VSMC and pericytes but not in cardiomyocytes, suggesting that a perturbation of Notch signalling in VSMC and pericytes indirectly impairs the cardiomyocytes. In keeping with this, Pdgfbret/ret mice, characterized by reduced numbers of VSMC and pericytes, showed left ventricular and cardiomyocyte hypertrophy. In conclusion, we demonstrate that reduced Notch3 or PDGFB signalling in vascular mural cells leads to cardiomyocyte dysfunction.
Collapse
Affiliation(s)
- Francesca Del Gaudio
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden.
| | - Dongli Liu
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
- Department of Pediatrics at the First Affiliated Hospital, Guangxi Medical University in Nanning, Guangxi, People's Republic of China
| | - Maarja Andaloussi Mäe
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Eike-Benjamin Braune
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Emil M Hansson
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Qing-Dong Wang
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Christer Betsholtz
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
- Department of Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Urban Lendahl
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
21
|
Weng T, Yang M, Zhang W, Jin R, Xia S, Zhang M, Wu P, He X, Han C, Zhao X, Wang X. Dual gene-activated dermal scaffolds regulate angiogenesis and wound healing by mediating the coexpression of VEGF and angiopoietin-1. Bioeng Transl Med 2023; 8:e10562. [PMID: 37693053 PMCID: PMC10487340 DOI: 10.1002/btm2.10562] [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: 06/30/2022] [Revised: 05/22/2023] [Accepted: 06/01/2023] [Indexed: 09/12/2023] Open
Abstract
The vascularization of dermal substitutes is a key challenge in efforts to heal deep skin defects. In this study, dual gene-activated dermal scaffolds (DGADSs-1) were fabricated by loading nanocomposite particles of polyethylenimine (PEI)/multiple plasmid DNAs (pDNAs) encoding vascular endothelial growth factor and angiopoietin-1 at a ratio of 1:1. In a similar manner, DGADSs-2 were loaded with a chimeric plasmid encoding both VEGF and Ang-1. In vitro studies showed that both types of DGADSs released PEI/pDNA nanoparticles in a sustained manner; they demonstrated effective transfection ability, leading to upregulated expression of VEGF and Ang-1. Furthermore, both types of DGADSs promoted fibroblast proliferation and blood vessel formation, although DGADSs-1 showed a more obvious promotion effect. A rat full-thickness skin defect model showed that split-thickness skin transplanted using a one-step method could achieve full survival at the 12th day after surgery in both DGADSs-1 and DGADSs-2 groups, and the vascularization time of dermal substitutes was significantly shortened. Compared with the other three groups of scaffolds, the DGADSs-1 group had significantly greater cell infiltration, collagen deposition, neovascularization, and vascular maturation, all of which promoted wound healing. Thus, compared with single-gene-activated dermal scaffolds, DGADSs show greater potential for enhancing angiogenesis. DGADSs with different loading modes also exhibited differences in terms of angiogenesis; the effect of loading two genes (DGADSs-1) was better than the effect of loading a chimeric gene (DGADSs-2). In summary, DGADSs, which continuously upregulate VEGF and Ang-1 expression, offer a new functional tissue-engineered dermal substitute with the ability to activate vascularization.
Collapse
Affiliation(s)
- Tingting Weng
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
- Department of Burn and Plastic SurgeryChildren's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical CenterHangzhouChina
| | - Min Yang
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| | - Wei Zhang
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| | - Ronghua Jin
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| | - Sizhan Xia
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| | - Manjia Zhang
- The First Clinical Medical College, Zhejiang Chinese Medical UniversityHangzhouChina
| | - Pan Wu
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| | - Xiaojie He
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| | - Chunmao Han
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| | - Xiong Zhao
- Department of Burn and Plastic SurgeryChildren's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical CenterHangzhouChina
| | - Xingang Wang
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| |
Collapse
|
22
|
Tatsukawa T, Kano K, Nakajima KI, Yazawa T, Eguchi R, Kabara M, Horiuchi K, Hayasaka T, Matsuo R, Hasebe N, Azuma N, Kawabe JI. NG2-positive pericytes regulate homeostatic maintenance of slow-type skeletal muscle with rapid myonuclear turnover. Stem Cell Res Ther 2023; 14:205. [PMID: 37592340 PMCID: PMC10433572 DOI: 10.1186/s13287-023-03433-1] [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: 01/24/2023] [Accepted: 07/26/2023] [Indexed: 08/19/2023] Open
Abstract
BACKGROUND Skeletal muscle comprises almost 40% of the human body and is essential for movement, structural support and metabolic homeostasis. Size of multinuclear skeletal muscle is stably maintained under steady conditions with the sporadic fusion of newly produced myocytes to compensate for the muscular turnover caused by daily wear and tear. It is becoming clear that microvascular pericytes (PCs) exhibit myogenic activity. However, whether PCs act as myogenic stem cells for the homeostatic maintenance of skeletal muscles during adulthood remains uncertain. METHODS We utilized PC-fused myofibers using PC-specific lineage tracing mouse (NG2-CreERT/Rosa-tdTomato) to observe whether muscle resident PCs have myogenic potential during daily life. Genetic PC deletion mouse model (NG2-CreERT/DTA) was used to test whether PC differentiates to myofibers for maintenance of muscle structure and function under homeostatic condition. RESULTS Under steady breeding conditions, tdTomato-expressing PCs were infused into myofibers, and subsequently, PC-derived nuclei were incorporated into myofibers. Especially in type-I slow-type myofibers such as the soleus, tdTomato+ myofibers were already observed 3 days after PC labeling; their ratio reached a peak (approximately 80%) within 1 month and was maintained for more than 1 year. Consistently, the NG2+ PC-specific deletion induced muscular atrophy in a slow-type myofiber-specific manner under steady breeding conditions. The number of myonucleus per volume of each myofiber was constant during observation period. CONCLUSIONS These findings demonstrate that the turnover of myonuclei in slow-type myofibers is relatively fast, with PCs acting as myogenic stem cells-the suppliers of new myonuclei under steady conditions-and play a vital role in the homeostatic maintenance of slow-type muscles.
Collapse
Affiliation(s)
- Takamitsu Tatsukawa
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
- Department of Vascular Surgery, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Kohei Kano
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
- Department of Cardiovascular Regeneration and Innovation, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Kei-Ichi Nakajima
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Takashi Yazawa
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Ryoji Eguchi
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Maki Kabara
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Kiwamu Horiuchi
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
- Division of Cardiovascular, Respiratory and Neurology, Department of Medicine, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Taiki Hayasaka
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
- Division of Cardiovascular, Respiratory and Neurology, Department of Medicine, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Risa Matsuo
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
- Department of Dermatology, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Naoyuki Hasebe
- Department of Cardiovascular Regeneration and Innovation, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
- Division of Cardiovascular, Respiratory and Neurology, Department of Medicine, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Nobuyoshi Azuma
- Department of Vascular Surgery, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Jun-Ichi Kawabe
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan.
- Department of Cardiovascular Regeneration and Innovation, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan.
| |
Collapse
|
23
|
Nicosia A, Salamone M, Costa S, Ragusa MA, Ghersi G. Mimicking Molecular Pathways in the Design of Smart Hydrogels for the Design of Vascularized Engineered Tissues. Int J Mol Sci 2023; 24:12314. [PMID: 37569691 PMCID: PMC10418696 DOI: 10.3390/ijms241512314] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 07/21/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Biomaterials are pivotal in supporting and guiding vascularization for therapeutic applications. To design effective, bioactive biomaterials, understanding the cellular and molecular processes involved in angiogenesis and vasculogenesis is crucial. Biomaterial platforms can replicate the interactions between cells, the ECM, and the signaling molecules that trigger blood vessel formation. Hydrogels, with their soft and hydrated properties resembling natural tissues, are widely utilized; particularly synthetic hydrogels, known for their bio-inertness and precise control over cell-material interactions, are utilized. Naturally derived and synthetic hydrogel bases are tailored with specific mechanical properties, controlled for biodegradation, and enhanced for cell adhesion, appropriate biochemical signaling, and architectural features that facilitate the assembly and tubulogenesis of vascular cells. This comprehensive review showcases the latest advancements in hydrogel materials and innovative design modifications aimed at effectively guiding and supporting vascularization processes. Furthermore, by leveraging this knowledge, researchers can advance biomaterial design, which will enable precise support and guidance of vascularization processes and ultimately enhance tissue functionality and therapeutic outcomes.
Collapse
Affiliation(s)
- Aldo Nicosia
- Institute for Biomedical Research and Innovation-National Research Council (IRIB-CNR), Via Ugo la Malfa 153, 90146 Palermo, Italy;
| | - Monica Salamone
- Institute for Biomedical Research and Innovation-National Research Council (IRIB-CNR), Via Ugo la Malfa 153, 90146 Palermo, Italy;
| | - Salvatore Costa
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, Ed. 16, 90128 Palermo, Italy; (S.C.); (M.A.R.); (G.G.)
| | - Maria Antonietta Ragusa
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, Ed. 16, 90128 Palermo, Italy; (S.C.); (M.A.R.); (G.G.)
| | - Giulio Ghersi
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, Ed. 16, 90128 Palermo, Italy; (S.C.); (M.A.R.); (G.G.)
| |
Collapse
|
24
|
Xiao P, Zhang Y, Zeng Y, Yang D, Mo J, Zheng Z, Wang J, Zhang Y, Zhou Z, Zhong X, Yan W. Impaired angiogenesis in ageing: the central role of the extracellular matrix. J Transl Med 2023; 21:457. [PMID: 37434156 PMCID: PMC10334673 DOI: 10.1186/s12967-023-04315-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/30/2023] [Indexed: 07/13/2023] Open
Abstract
Each step in angiogenesis is regulated by the extracellular matrix (ECM). Accumulating evidence indicates that ageing-related changes in the ECM driven by cellular senescence lead to a reduction in neovascularisation, reduced microvascular density, and an increased risk of tissue ischaemic injury. These changes can lead to health events that have major negative impacts on quality of life and place a significant financial burden on the healthcare system. Elucidating interactions between the ECM and cells during angiogenesis in the context of ageing is neceary to clarify the mechanisms underlying reduced angiogenesis in older adults. In this review, we summarize ageing-related changes in the composition, structure, and function of the ECM and their relevance for angiogenesis. Then, we explore in detail the mechanisms of interaction between the aged ECM and cells during impaired angiogenesis in the older population for the first time, discussing diseases caused by restricted angiogenesis. We also outline several novel pro-angiogenic therapeutic strategies targeting the ECM that can provide new insights into the choice of appropriate treatments for a variety of age-related diseases. Based on the knowledge gathered from recent reports and journal articles, we provide a better understanding of the mechanisms underlying impaired angiogenesis with age and contribute to the development of effective treatments that will enhance quality of life.
Collapse
Affiliation(s)
- Ping Xiao
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yanli Zhang
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Yuting Zeng
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Dehong Yang
- Department of Orthopedics Spinal Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jiayao Mo
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ziting Zheng
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jilei Wang
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yuxin Zhang
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zhiyan Zhou
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xincen Zhong
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Wenjuan Yan
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
25
|
Liu G, Wang J, Wei Z, Fang C, Shen K, Qian C, Qi C, Li T, Gao P, Wong PC, Lu H, Cao X, Wan M. Elevated PDGF-BB from Bone Impairs Hippocampal Vasculature by Inducing PDGFRβ Shedding from Pericytes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206938. [PMID: 37102631 PMCID: PMC10369301 DOI: 10.1002/advs.202206938] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 04/11/2023] [Indexed: 06/19/2023]
Abstract
Evidence suggests a unique association between bone aging and neurodegenerative/cerebrovascular disorders. However, the mechanisms underlying bone-brain interplay remain elusive. Here platelet-derived growth factor-BB (PDGF-BB) produced by preosteoclasts in bone is reported to promote age-associated hippocampal vascular impairment. Aberrantly elevated circulating PDGF-BB in aged mice and high-fat diet (HFD)-challenged mice correlates with capillary reduction, pericyte loss, and increased blood-brain barrier (BBB) permeability in their hippocampus. Preosteoclast-specific Pdgfb transgenic mice with markedly high plasma PDGF-BB concentration faithfully recapitulate the age-associated hippocampal BBB impairment and cognitive decline. Conversely, preosteoclast-specific Pdgfb knockout mice have attenuated hippocampal BBB impairment in aged mice or HFD-challenged mice. Persistent exposure of brain pericytes to high concentrations of PDGF-BB upregulates matrix metalloproteinase 14 (MMP14), which promotes ectodomain shedding of PDGF receptor β (PDGFRβ) from pericyte surface. MMP inhibitor treatment alleviates hippocampal pericyte loss and capillary reduction in the conditional Pdgfb transgenic mice and antagonizes BBB leakage in aged mice. The findings establish the role of bone-derived PDGF-BB in mediating hippocampal BBB disruption and identify the ligand-induced PDGFRβ shedding as a feedback mechanism for age-associated PDGFRβ downregulation and the consequent pericyte loss.
Collapse
Affiliation(s)
- Guanqiao Liu
- Department of Orthopaedic SurgeryJohns Hopkins University School of MedicineRoss Building, Room 232, 720 Rutland AvenueBaltimoreMD21205USA
| | - Jiekang Wang
- Department of Orthopaedic SurgeryJohns Hopkins University School of MedicineRoss Building, Room 232, 720 Rutland AvenueBaltimoreMD21205USA
| | - Zhiliang Wei
- The Russell H. Morgan Department of Radiology and Radiological ScienceThe Johns Hopkins University School of MedicineBaltimoreMD21205USA
| | - Ching‐Lien Fang
- Department of Orthopaedic SurgeryJohns Hopkins University School of MedicineRoss Building, Room 232, 720 Rutland AvenueBaltimoreMD21205USA
| | - Ke Shen
- Department of Orthopaedic SurgeryJohns Hopkins University School of MedicineRoss Building, Room 232, 720 Rutland AvenueBaltimoreMD21205USA
| | - Cheng Qian
- Department of Orthopaedic SurgeryJohns Hopkins University School of MedicineRoss Building, Room 232, 720 Rutland AvenueBaltimoreMD21205USA
| | - Cheng Qi
- Department of Orthopaedic SurgeryJohns Hopkins University School of MedicineRoss Building, Room 232, 720 Rutland AvenueBaltimoreMD21205USA
| | - Tong Li
- Department of PathologyJohns Hopkins University School of MedicineBaltimoreMD21205USA
| | - Peisong Gao
- Division of Allergy and Clinical ImmunologyJohns Hopkins University School of MedicineBaltimoreMD21224USA
| | - Philip C. Wong
- Department of PathologyJohns Hopkins University School of MedicineBaltimoreMD21205USA
- Department of NeuroscienceJohns Hopkins University School of MedicineBaltimoreMD21205USA
| | - Hanzhang Lu
- The Russell H. Morgan Department of Radiology and Radiological ScienceThe Johns Hopkins University School of MedicineBaltimoreMD21205USA
| | - Xu Cao
- Department of Orthopaedic SurgeryJohns Hopkins University School of MedicineRoss Building, Room 232, 720 Rutland AvenueBaltimoreMD21205USA
| | - Mei Wan
- Department of Orthopaedic SurgeryJohns Hopkins University School of MedicineRoss Building, Room 232, 720 Rutland AvenueBaltimoreMD21205USA
| |
Collapse
|
26
|
Chen X, Wolfe DA, Bindu DS, Zhang M, Taskin N, Goertsen D, Shay TF, Sullivan EE, Huang SF, Ravindra Kumar S, Arokiaraj CM, Plattner VM, Campos LJ, Mich JK, Monet D, Ngo V, Ding X, Omstead V, Weed N, Bishaw Y, Gore BB, Lein ES, Akrami A, Miller C, Levi BP, Keller A, Ting JT, Fox AS, Eroglu C, Gradinaru V. Functional gene delivery to and across brain vasculature of systemic AAVs with endothelial-specific tropism in rodents and broad tropism in primates. Nat Commun 2023; 14:3345. [PMID: 37291094 PMCID: PMC10250345 DOI: 10.1038/s41467-023-38582-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 05/02/2023] [Indexed: 06/10/2023] Open
Abstract
Delivering genes to and across the brain vasculature efficiently and specifically across species remains a critical challenge for addressing neurological diseases. We have evolved adeno-associated virus (AAV9) capsids into vectors that transduce brain endothelial cells specifically and efficiently following systemic administration in wild-type mice with diverse genetic backgrounds, and in rats. These AAVs also exhibit superior transduction of the CNS across non-human primates (marmosets and rhesus macaques), and in ex vivo human brain slices, although the endothelial tropism is not conserved across species. The capsid modifications translate from AAV9 to other serotypes such as AAV1 and AAV-DJ, enabling serotype switching for sequential AAV administration in mice. We demonstrate that the endothelial-specific mouse capsids can be used to genetically engineer the blood-brain barrier by transforming the mouse brain vasculature into a functional biofactory. We apply this approach to Hevin knockout mice, where AAV-X1-mediated ectopic expression of the synaptogenic protein Sparcl1/Hevin in brain endothelial cells rescued synaptic deficits.
Collapse
Grants
- P51 OD010425 NIH HHS
- P51 OD011107 NIH HHS
- Howard Hughes Medical Institute
- DP1 NS111369 NINDS NIH HHS
- OT2 OD024899 NIH HHS
- DP1 MH104069 NIMH NIH HHS
- UF1 MH128336 NIMH NIH HHS
- DP1 EB016986 NIBIB NIH HHS
- DP1 OD000616 NIH HHS
- DP2 NS087949 NINDS NIH HHS
- UG3 MH120095 NIMH NIH HHS
- U42 OD011123 NIH HHS
- NIH Director’s New Innovator DP2NS087949 and PECASE, NIH BRAIN Armamentarium 1UF1MH128336-01, NIH Pioneer 5DP1NS111369-04 and SPARC 1OT2OD024899. Additional funding includes the Vallee Foundation, the Moore Foundation, the CZI Neurodegeneration Challenge Network, and the NSF NeuroNex Technology Hub grant 1707316, the Heritage Medical Research Institute and the Beckman Institute for CLARITY, Optogenetics and Vector Engineering Research (CLOVER) for technology development and dissemination, NIH BRAIN UG3MH120095.
- The Swiss National Science Foundation (310030_188952, A.K), the Synapsis (grant 2019-PI02, A.K.), the Swiss Multiple Sclerosis Society (A.K.).
- CNPRC base grant (NIH P51 OD011107)
- The CZI Neurodegeneration Challenge Network. C.E. is an investigator of the Howard Hughes Medical Institute.
Collapse
Affiliation(s)
- Xinhong Chen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Damien A Wolfe
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | | | - Mengying Zhang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Naz Taskin
- Allen Institute for Brain Science, Seattle, WA, USA
| | - David Goertsen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Timothy F Shay
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Erin E Sullivan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Sheng-Fu Huang
- Department of Neurosurgery, Clinical Neuroscience Center, Zürich University Hospital, University of Zürich, Zürich, Switzerland
| | - Sripriya Ravindra Kumar
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Cynthia M Arokiaraj
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | | | - Lillian J Campos
- Department of Psychology and California National Primate Research Center, University of California, Davis, Davis, CA, 95616, USA
| | - John K Mich
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Deja Monet
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Victoria Ngo
- Cortical Systems and Behavior Lab, University of California San Diego, La Jolla, CA, 92039, USA
| | - Xiaozhe Ding
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | | | - Natalie Weed
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Yeme Bishaw
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Bryan B Gore
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Ed S Lein
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Athena Akrami
- Sainsbury Wellcome Centre, University College London, London, UK
| | - Cory Miller
- Cortical Systems and Behavior Lab, University of California San Diego, La Jolla, CA, 92039, USA
| | - Boaz P Levi
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Annika Keller
- Department of Neurosurgery, Clinical Neuroscience Center, Zürich University Hospital, University of Zürich, Zürich, Switzerland
- Neuroscience Center Zürich, University of Zürich and ETH Zürich, Zürich, Switzerland
| | - Jonathan T Ting
- Allen Institute for Brain Science, Seattle, WA, USA
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Andrew S Fox
- Department of Psychology and California National Primate Research Center, University of California, Davis, Davis, CA, 95616, USA
| | - Cagla Eroglu
- Department of Cell Biology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA.
| |
Collapse
|
27
|
Yasuda K, Noma H, Mimura T, Nonaka R, Sasaki S, Suganuma N, Shimura M. Effects of Intravitreal Ranibizumab Injection on Peripheral Retinal Microcirculation and Cytokines in Branch Retinal Vein Occlusion with Macular Edema. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1053. [PMID: 37374257 DOI: 10.3390/medicina59061053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/26/2023] [Accepted: 05/28/2023] [Indexed: 06/29/2023]
Abstract
Background and Objectives: To investigate peripheral blood flow in retinal vessels and vessel diameters after intravitreal ranibizumab injection (IRI) and the relationship between these parameters and cytokines in branch retinal vein occlusion (BRVO) with macular edema. Materials and Methods: We assessed relative flow volume (RFV) and the width of the main and branch retinal arteries and veins in the occluded and non-occluded regions before and after IRI in 37 patients with BRVO and macular edema. Measurements were made using laser speckle flowgraphy (LSFG). When performing IRI, we obtained samples of aqueous humor and analyzed them using the suspension array method to evaluate vascular endothelial growth factor (VEGF), placental growth factor (PlGF), platelet-derived growth factor (PDGF)-AA, soluble intercellular adhesion molecule (sICAM)-1, monocyte chemoattractant protein 1 (MCP-1), interleukin (IL)-6, IL-8, and interferon-inducible 10-kDa protein (IP-10). Results: In both retinal regions, before and after IRI, the RFV in the main artery and vein showed a significant correlation with the summed RFV in the respective branch vessels 1 and 2. In the occluded region, the RFV in the main vein was significantly negatively correlated with MCP-1, PDGF-AA, IL-6, and IL-8; the RFV in branch vein 1 was significantly negatively correlated with PlGF, MCP-1, IL-6, and IL-8; PDGF-AA was significantly negatively correlated with the width of the main and branch veins; and the RFVs of the main artery and vein decreased significantly from before to 1 month after IRI. Conclusions: Contrary to expectations, the study found that anti-VEGF therapy does not affect RFV in arteries and veins in patients with BRVO and macular edema. Furthermore, retinal blood flow is poor in patients with high MCP-1, IL-6, and IL-8. Finally, high PDGF-AA may result in smaller venous diameters and reduced retinal blood flow.
Collapse
Affiliation(s)
- Kanako Yasuda
- Department of Ophthalmology, Hachioji Medical Center, Tokyo Medical University, 1163, Tatemachi, Hachioji, Tokyo 193-0998, Japan
| | - Hidetaka Noma
- Department of Ophthalmology, Hachioji Medical Center, Tokyo Medical University, 1163, Tatemachi, Hachioji, Tokyo 193-0998, Japan
| | - Tatsuya Mimura
- Department of Ophthalmology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8606, Japan
| | - Ryota Nonaka
- Department of Ophthalmology, Hachioji Medical Center, Tokyo Medical University, 1163, Tatemachi, Hachioji, Tokyo 193-0998, Japan
| | - Shotaro Sasaki
- Department of Ophthalmology, Hachioji Medical Center, Tokyo Medical University, 1163, Tatemachi, Hachioji, Tokyo 193-0998, Japan
| | - Noboru Suganuma
- Department of Ophthalmology, Hachioji Medical Center, Tokyo Medical University, 1163, Tatemachi, Hachioji, Tokyo 193-0998, Japan
| | - Masahiko Shimura
- Department of Ophthalmology, Hachioji Medical Center, Tokyo Medical University, 1163, Tatemachi, Hachioji, Tokyo 193-0998, Japan
| |
Collapse
|
28
|
Orozco-García E, van Meurs DJ, Calderón JC, Narvaez-Sanchez R, Harmsen MC. Endothelial plasticity across PTEN and Hippo pathways: A complex hormetic rheostat modulated by extracellular vesicles. Transl Oncol 2023; 31:101633. [PMID: 36905871 PMCID: PMC10020115 DOI: 10.1016/j.tranon.2023.101633] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/20/2022] [Accepted: 01/25/2023] [Indexed: 03/11/2023] Open
Abstract
Vascularization is a multifactorial and spatiotemporally regulated process, essential for cell and tissue survival. Vascular alterations have repercussions on the development and progression of diseases such as cancer, cardiovascular diseases, and diabetes, which are the leading causes of death worldwide. Additionally, vascularization continues to be a challenge for tissue engineering and regenerative medicine. Hence, vascularization is the center of interest for physiology, pathophysiology, and therapeutic processes. Within vascularization, phosphatase and tensin homolog deleted on chromosome 10 (PTEN) and Hippo signaling have pivotal roles in the development and homeostasis of the vascular system. Their suppression is related to several pathologies, including developmental defects and cancer. Non-coding RNAs (ncRNAs) are among the regulators of PTEN and/or Hippo pathways during development and disease. The purpose of this paper is to review and discuss the mechanisms by which exosome-derived ncRNAs modulate endothelial cell plasticity during physiological and pathological angiogenesis, through the regulation of PTEN and Hippo pathways, aiming to establish new perspectives on cellular communication during tumoral and regenerative vascularization.
Collapse
Affiliation(s)
- Elizabeth Orozco-García
- Physiology and biochemistry research group - PHYSIS, Faculty of Medicine, University of Antioquia, Colombia; Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1 (EA11), Groningen 9713 GZ, The Netherlands
| | - D J van Meurs
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1 (EA11), Groningen 9713 GZ, The Netherlands
| | - J C Calderón
- Physiology and biochemistry research group - PHYSIS, Faculty of Medicine, University of Antioquia, Colombia
| | - Raul Narvaez-Sanchez
- Physiology and biochemistry research group - PHYSIS, Faculty of Medicine, University of Antioquia, Colombia
| | - M C Harmsen
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1 (EA11), Groningen 9713 GZ, The Netherlands.
| |
Collapse
|
29
|
Payne LB, Abdelazim H, Hoque M, Barnes A, Mironovova Z, Willi CE, Darden J, Houk C, Sedovy MW, Johnstone SR, Chappell JC. A Soluble Platelet-Derived Growth Factor Receptor-β Originates via Pre-mRNA Splicing in the Healthy Brain and Is Upregulated during Hypoxia and Aging. Biomolecules 2023; 13:711. [PMID: 37189457 PMCID: PMC10136073 DOI: 10.3390/biom13040711] [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: 01/19/2023] [Revised: 04/17/2023] [Accepted: 04/19/2023] [Indexed: 05/17/2023] Open
Abstract
The platelet-derived growth factor-BB (PDGF-BB) pathway provides critical regulation of cerebrovascular pericytes, orchestrating their investment and retention within the brain microcirculation. Dysregulated PDGF Receptor-beta (PDGFRβ) signaling can lead to pericyte defects that compromise blood-brain barrier (BBB) integrity and cerebral perfusion, impairing neuronal activity and viability, which fuels cognitive and memory deficits. Receptor tyrosine kinases such as PDGF-BB and vascular endothelial growth factor-A (VEGF-A) are often modulated by soluble isoforms of cognate receptors that establish signaling activity within a physiological range. Soluble PDGFRβ (sPDGFRβ) isoforms have been reported to form by enzymatic cleavage from cerebrovascular mural cells, and pericytes in particular, largely under pathological conditions. However, pre-mRNA alternative splicing has not been widely explored as a possible mechanism for generating sPDGFRβ variants, and specifically during tissue homeostasis. Here, we found sPDGFRβ protein in the murine brain and other tissues under normal, physiological conditions. Utilizing brain samples for follow-on analysis, we identified mRNA sequences corresponding to sPDGFRβ isoforms, which facilitated construction of predicted protein structures and related amino acid sequences. Human cell lines yielded comparable sequences and protein model predictions. Retention of ligand binding capacity was confirmed for sPDGFRβ by co-immunoprecipitation. Visualizing fluorescently labeled sPDGFRβ transcripts revealed a spatial distribution corresponding to murine brain pericytes alongside cerebrovascular endothelium. Soluble PDGFRβ protein was detected throughout the brain parenchyma in distinct regions, such as along the lateral ventricles, with signals also found more broadly adjacent to cerebral microvessels consistent with pericyte labeling. To better understand how sPDGFRβ variants might be regulated, we found elevated transcript and protein levels in the murine brain with age, and acute hypoxia increased sPDGFRβ variant transcripts in a cell-based model of intact vessels. Our findings indicate that soluble isoforms of PDGFRβ likely arise from pre-mRNA alternative splicing, in addition to enzymatic cleavage mechanisms, and these variants exist under normal physiological conditions. Follow-on studies will be needed to establish potential roles for sPDGFRβ in regulating PDGF-BB signaling to maintain pericyte quiescence, BBB integrity, and cerebral perfusion-critical processes underlying neuronal health and function, and in turn, memory and cognition.
Collapse
Affiliation(s)
- Laura Beth Payne
- Fralin Biomedical Research Institute (FBRI) at Virginia Tech-Carilion (VTC), Roanoke, VA 24016, USA
- FBRI Center for Vascular and Heart Research, Roanoke, VA 24016, USA
| | - Hanaa Abdelazim
- Fralin Biomedical Research Institute (FBRI) at Virginia Tech-Carilion (VTC), Roanoke, VA 24016, USA
- FBRI Center for Vascular and Heart Research, Roanoke, VA 24016, USA
| | - Maruf Hoque
- Fralin Biomedical Research Institute (FBRI) at Virginia Tech-Carilion (VTC), Roanoke, VA 24016, USA
- FBRI Center for Vascular and Heart Research, Roanoke, VA 24016, USA
| | - Audra Barnes
- Fralin Biomedical Research Institute (FBRI) at Virginia Tech-Carilion (VTC), Roanoke, VA 24016, USA
- FBRI Center for Vascular and Heart Research, Roanoke, VA 24016, USA
- Department of Biomedical Engineering and Mechanics, School of Biomedical Engineering and Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| | - Zuzana Mironovova
- Fralin Biomedical Research Institute (FBRI) at Virginia Tech-Carilion (VTC), Roanoke, VA 24016, USA
- FBRI Center for Vascular and Heart Research, Roanoke, VA 24016, USA
| | - Caroline E. Willi
- Fralin Biomedical Research Institute (FBRI) at Virginia Tech-Carilion (VTC), Roanoke, VA 24016, USA
- FBRI Center for Vascular and Heart Research, Roanoke, VA 24016, USA
| | - Jordan Darden
- Fralin Biomedical Research Institute (FBRI) at Virginia Tech-Carilion (VTC), Roanoke, VA 24016, USA
- FBRI Center for Vascular and Heart Research, Roanoke, VA 24016, USA
| | - Clifton Houk
- Fralin Biomedical Research Institute (FBRI) at Virginia Tech-Carilion (VTC), Roanoke, VA 24016, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
| | - Meghan W. Sedovy
- Fralin Biomedical Research Institute (FBRI) at Virginia Tech-Carilion (VTC), Roanoke, VA 24016, USA
- FBRI Center for Vascular and Heart Research, Roanoke, VA 24016, USA
| | - Scott R. Johnstone
- Fralin Biomedical Research Institute (FBRI) at Virginia Tech-Carilion (VTC), Roanoke, VA 24016, USA
- FBRI Center for Vascular and Heart Research, Roanoke, VA 24016, USA
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| | - John C. Chappell
- Fralin Biomedical Research Institute (FBRI) at Virginia Tech-Carilion (VTC), Roanoke, VA 24016, USA
- FBRI Center for Vascular and Heart Research, Roanoke, VA 24016, USA
- Department of Biomedical Engineering and Mechanics, School of Biomedical Engineering and Sciences, Virginia Tech, Blacksburg, VA 24061, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
| |
Collapse
|
30
|
Zhu W, Huang X. Mural cell composition and functional analysis in the healing process of human gingiva from periodontal intrabony defects. Arch Oral Biol 2023; 150:105687. [PMID: 36947913 DOI: 10.1016/j.archoralbio.2023.105687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/23/2023] [Accepted: 03/15/2023] [Indexed: 03/19/2023]
Abstract
OBJECTIVE To evaluate the composition and function of mural cell populations in human gingival tissues DESIGN: A cross-sectional study was conducted on seven periodontitis (stage Ⅲ) patients. Gingival tissues were collected two months after scaling and root planing and divided into 3 groups: 1, h_h group (horizontal bone resorption, residual pocket depth ≤3 mm); 2, v_h group (vertical bone resorption >4 mm, residual pocket depth ≤3 mm); 3, v_i group (vertical bone resorption >4 mm, residual pocket depth ≥6 mm). Single-cell RNA sequencing (10X genomics) and subsequent bioinformatics analysis were performed. Protein expression of selected genes was confirmed by histological staining. RESULTS Two mural cell clusters, RGS5+THY1+ and ACTA2+MYH11+ subpopulations, were identified and confirmed by histological staining and cross-validation with three different single-cell RNA sequencing datasets in the GEO database. RGS5+THY1+ cluster in perivascular areas possessed cellular protrusions and exhibited immunomodulatory and synthetic phenotypes. In contrast, the ACTA2+MYH11+ cluster strictly distributed around vessel walls was characterized by a contractile phenotype. Mural cells closely interacted with endothelial cells through PDGF and NOTCH3 signaling. Mural cell loss was detected in the v_i group and in hopeless periodontal teeth, which might be caused by tumor necrosis factor-alpha induced apoptosis. CONCLUSIONS Gingival mural cells can be classified into two distinct clusters according to their gene signatures and cell morphology. The loss of mural cells may indicate periodontitis progression.
Collapse
Affiliation(s)
- Wenjun Zhu
- Department of Periodontology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China.
| | - Xin Huang
- Department of Periodontology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| |
Collapse
|
31
|
The Active Role of Pericytes During Neuroinflammation in the Adult Brain. Cell Mol Neurobiol 2023; 43:525-541. [PMID: 35195811 DOI: 10.1007/s10571-022-01208-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 02/13/2022] [Indexed: 12/11/2022]
Abstract
Microvessels in the central nervous system (CNS) have one of the highest populations of pericytes, indicating their crucial role in maintaining homeostasis. Pericytes are heterogeneous cells located around brain microvessels; they present three different morphologies along the CNS vascular tree: ensheathing, mesh, and thin-strand pericytes. At the arteriole-capillary transition ensheathing pericytes are found, while mesh and thin-strand pericytes are located at capillary beds. Brain pericytes are essential for the establishment and maintenance of the blood-brain barrier, which restricts the passage of soluble and potentially toxic molecules from the circulatory system to the brain parenchyma. Pericytes play a key role in regulating local inflammation at the CNS. Pericytes can respond differentially, depending on the degree of inflammation, by secreting a set of neurotrophic factors to promote cell survival and regeneration, or by potentiating inflammation through the release of inflammatory mediators (e.g., cytokines and chemokines), and the overexpression of cell adhesion molecules. Under inflammatory conditions, pericytes may regulate immune cell trafficking to the CNS and play a role in perpetuating local inflammation. In this review, we describe pericyte responses during acute and chronic neuroinflammation.
Collapse
|
32
|
Payne LB, Abdelazim H, Hoque M, Barnes A, Mironovova Z, Willi CE, Darden J, Jenkins-Houk C, Sedovy MW, Johnstone SR, Chappell JC. A Soluble Platelet-Derived Growth Factor Receptor-β Originates via Pre-mRNA Splicing in the Healthy Brain and is Differentially Regulated during Hypoxia and Aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.03.527005. [PMID: 36778261 PMCID: PMC9915746 DOI: 10.1101/2023.02.03.527005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The platelet-derived growth factor-BB (PDGF-BB) pathway provides critical regulation of cerebrovascular pericytes, orchestrating their investment and retention within the brain microcirculation. Dysregulated PDGF Receptor-beta (PDGFRβ) signaling can lead to pericyte defects that compromise blood-brain barrier (BBB) integrity and cerebral perfusion, impairing neuronal activity and viability, which fuels cognitive and memory deficits. Receptor tyrosine kinases (RTKs) like PDGF-BB and vascular endothelial growth factor-A (VEGF-A) are often modulated by soluble isoforms of cognate receptors that establish signaling activity within a physiological range. Soluble PDGFRβ (sPDGFRβ) isoforms have been reported to form by enzymatic cleavage from cerebrovascular mural cells, and pericytes in particular, largely under pathological conditions. However, pre-mRNA alternative splicing has not been widely explored as a possible mechanism for generating sPDGFRβ variants, and specifically during tissue homeostasis. Here, we found sPDGFRβ protein in the murine brain and other tissues under normal, physiological conditions. Utilizing brain samples for follow-on analysis, we identified mRNA sequences corresponding to sPDGFRβ isoforms, which facilitated construction of predicted protein structures and related amino acid sequences. Human cell lines yielded comparable sequences and protein model predictions. Retention of ligand binding capacity was confirmed for sPDGFRβ by co-immunoprecipitation. Visualizing fluorescently labeled sPDGFRβ transcripts revealed a spatial distribution corresponding to murine brain pericytes alongside cerebrovascular endothelium. Soluble PDGFRβ protein was detected throughout the brain parenchyma in distinct regions such as along the lateral ventricles, with signals also found more broadly adjacent to cerebral microvessels consistent with pericyte labeling. To better understand how sPDGFRβ variants might be regulated, we found elevated transcript and protein levels in the murine brain with age, and acute hypoxia increased sPDGFRβ variant transcripts in a cell-based model of intact vessels. Our findings indicate that soluble isoforms of PDGFRβ likely arise from pre-mRNA alternative splicing, in addition to enzymatic cleavage mechanisms, and these variants exist under normal physiological conditions. Follow-on studies will be needed to establish potential roles for sPDGFRβ in regulating PDGF-BB signaling to maintain pericyte quiescence, BBB integrity, and cerebral perfusion - critical processes underlying neuronal health and function, and in turn memory and cognition.
Collapse
|
33
|
Zhang Y, Ren Y, Zhang Y, Li Y, Xu C, Peng Z, Jia Y, Qiao S, Zhang Z, Shi L. T-cell infiltration in the central nervous system and their association with brain calcification in Slc20a2-deficient mice. Front Mol Neurosci 2023; 16:1073723. [PMID: 36741925 PMCID: PMC9894888 DOI: 10.3389/fnmol.2023.1073723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 01/03/2023] [Indexed: 01/21/2023] Open
Abstract
Primary familial brain calcification (PFBC) is a rare neurodegenerative and neuropsychiatric disorder characterized by bilateral symmetric intracranial calcification along the microvessels or inside neuronal cells in the basal ganglia, thalamus, and cerebellum. Slc20a2 homozygous (HO) knockout mice are the most commonly used model to simulate the brain calcification phenotype observed in human patients. However, the cellular and molecular mechanisms related to brain calcification, particularly at the early stage much prior to the emergence of brain calcification, remain largely unknown. In this study, we quantified the central nervous system (CNS)-infiltrating T-cells of different age groups of Slc20a2-HO and matched wild type mice and found CD45+CD3+ T-cells to be significantly increased in the brain parenchyma, even in the pre-calcification stage of 1-month-old -HO mice. The accumulation of the CD3+ T-cells appeared to be associated with the severity of brain calcification. Further immunophenotyping revealed that the two main subtypes that had increased in the brain were CD3+ CD4- CD8- and CD3+ CD4+ T-cells. The expression of endothelial cell (EC) adhesion molecules increased, while that of tight and adherents junction proteins decreased, providing the molecular precondition for T-cell recruitment to ECs and paracellular migration into the brain. The fusion of lymphocytes and EC membranes and transcellular migration of CD3-related gold particles were captured, suggesting enhancement of transcytosis in the brain ECs. Exogenous fluorescent tracers and endogenous IgG and albumin leakage also revealed an impairment of transcellular pathway in the ECs. FTY720 significantly alleviated brain calcification, probably by reducing T-cell infiltration, modulating neuroinflammation and ossification process, and enhancing the autophagy and phagocytosis of CNS-resident immune cells. This study clearly demonstrated CNS-infiltrating T-cells to be associated with the progression of brain calcification. Impairment of blood-brain barrier (BBB) permeability, which was closely related to T-cell invasion into the CNS, could be explained by the BBB alterations of an increase in the paracellular and transcellular pathways of brain ECs. FTY720 was found to be a potential drug to protect patients from PFBC-related lesions in the future.
Collapse
Affiliation(s)
- Yi Zhang
- Human Molecular Genetics Group, NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China,Department of Medical Genetics, College of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Yaqiong Ren
- Human Molecular Genetics Group, NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yueni Zhang
- Human Molecular Genetics Group, NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China,Department of Medical Genetics, College of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Ying Li
- Human Molecular Genetics Group, NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China,Department of Child and Adolescent Health, School of Public Health, Harbin Medical University, Harbin, China
| | - Chao Xu
- Human Molecular Genetics Group, NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China,Department of Pediatrics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ziyue Peng
- Human Molecular Genetics Group, NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China,Department of Pediatrics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ying Jia
- Department of Medical Genetics, College of Basic Medical Sciences, Harbin Medical University, Harbin, China,Department of Child and Adolescent Health, School of Public Health, Harbin Medical University, Harbin, China
| | - Shupei Qiao
- Human Molecular Genetics Group, NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China,Department of Child and Adolescent Health, School of Public Health, Harbin Medical University, Harbin, China
| | - Zitong Zhang
- Human Molecular Genetics Group, NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China,Department of Medical Genetics, College of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Lei Shi
- Human Molecular Genetics Group, NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China,Department of Medical Genetics, College of Basic Medical Sciences, Harbin Medical University, Harbin, China,*Correspondence: Lei Shi,
| |
Collapse
|
34
|
Chen X, Wolfe DA, Bindu DS, Zhang M, Taskin N, Goertsen D, Shay TF, Sullivan E, Huang SF, Kumar SR, Arokiaraj CM, Plattner V, Campos LJ, Mich J, Monet D, Ngo V, Ding X, Omstead V, Weed N, Bishaw Y, Gore B, Lein ES, Akrami A, Miller C, Levi BP, Keller A, Ting JT, Fox AS, Eroglu C, Gradinaru V. Functional gene delivery to and across brain vasculature of systemic AAVs with endothelial-specific tropism in rodents and broad tropism in primates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.12.523844. [PMID: 36711773 PMCID: PMC9882234 DOI: 10.1101/2023.01.12.523844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Delivering genes to and across the brain vasculature efficiently and specifically across species remains a critical challenge for addressing neurological diseases. We have evolved adeno-associated virus (AAV9) capsids into vectors that transduce brain endothelial cells specifically and efficiently following systemic administration in wild-type mice with diverse genetic backgrounds and rats. These AAVs also exhibit superior transduction of the CNS across non-human primates (marmosets and rhesus macaques), and ex vivo human brain slices although the endothelial tropism is not conserved across species. The capsid modifications translate from AAV9 to other serotypes such as AAV1 and AAV-DJ, enabling serotype switching for sequential AAV administration in mice. We demonstrate that the endothelial specific mouse capsids can be used to genetically engineer the blood-brain barrier by transforming the mouse brain vasculature into a functional biofactory. Vasculature-secreted Hevin (a synaptogenic protein) rescued synaptic deficits in a mouse model.
Collapse
Affiliation(s)
- Xinhong Chen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Damien A Wolfe
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | | | - Mengying Zhang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Naz Taskin
- Allen Institute for Brain Science, Seattle, WA, USA
| | - David Goertsen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Timothy F Shay
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Erin Sullivan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Sheng-Fu Huang
- Department of Neurosurgery, Clinical Neuroscience Center, Zurich University Hospital, University of Zurich, Zurich, Switzerland
| | - Sripriya Ravindra Kumar
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Cynthia M Arokiaraj
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Viktor Plattner
- Sainsbury Wellcome Centre, University College London, London, UK
| | - Lillian J Campos
- Department of Psychology and California National Primate Research Center, University of California, Davis, Davis, CA, 95616, USA
| | - John Mich
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Deja Monet
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Victoria Ngo
- Cortical Systems and Behavior Lab, University of California San Diego, La Jolla, CA, 92039, USA
| | - Xiaozhe Ding
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | | | - Natalie Weed
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Yeme Bishaw
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Bryan Gore
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Ed S Lein
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Athena Akrami
- Sainsbury Wellcome Centre, University College London, London, UK
| | - Cory Miller
- Cortical Systems and Behavior Lab, University of California San Diego, La Jolla, CA, 92039, USA
| | - Boaz P Levi
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Annika Keller
- Department of Neurosurgery, Clinical Neuroscience Center, Zurich University Hospital, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Jonathan T Ting
- Allen Institute for Brain Science, Seattle, WA, USA
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Andrew S Fox
- Department of Psychology and California National Primate Research Center, University of California, Davis, Davis, CA, 95616, USA
| | - Cagla Eroglu
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| |
Collapse
|
35
|
Zeng J, Peng Y, Wang D, Ayesha K, Chen S. The interaction between osteosarcoma and other cells in the bone microenvironment: From mechanism to clinical applications. Front Cell Dev Biol 2023; 11:1123065. [PMID: 37206921 PMCID: PMC10189553 DOI: 10.3389/fcell.2023.1123065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 04/10/2023] [Indexed: 05/21/2023] Open
Abstract
Osteosarcoma is a primary bone tumor with a high mortality rate. The event-free survival rate has not improved significantly in the past 30 years, which brings a heavy burden to patients and society. The high heterogeneity of osteosarcoma leads to the lack of specific targets and poor therapeutic effect. Tumor microenvironment is the focus of current research, and osteosarcoma is closely related to bone microenvironment. Many soluble factors and extracellular matrix secreted by many cells in the bone microenvironment have been shown to affect the occurrence, proliferation, invasion and metastasis of osteosarcoma through a variety of signaling pathways. Therefore, targeting other cells in the bone microenvironment may improve the prognosis of osteosarcoma. The mechanism by which osteosarcoma interacts with other cells in the bone microenvironment has been extensively investigated, but currently developed drugs targeting the bone microenvironment have poor efficacy. Therefore, we review the regulatory effects of major cells and physical and chemical properties in the bone microenvironment on osteosarcoma, focusing on their complex interactions, potential therapeutic strategies and clinical applications, to deepen our understanding of osteosarcoma and the bone microenvironment and provide reference for future treatment. Targeting other cells in the bone microenvironment may provide potential targets for the development of clinical drugs for osteosarcoma and may improve the prognosis of osteosarcoma.
Collapse
Affiliation(s)
- Jin Zeng
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yi Peng
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Dong Wang
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Khan Ayesha
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Shijie Chen
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
- *Correspondence: Shijie Chen,
| |
Collapse
|
36
|
Stobart JL, Erlebach E, Glück C, Huang SF, Barrett MJ, Li M, Vinogradov SA, Klohs J, Zarb Y, Keller A, Weber B. Altered hemodynamics and vascular reactivity in a mouse model with severe pericyte deficiency. J Cereb Blood Flow Metab 2022; 43:763-777. [PMID: 36545806 PMCID: PMC10108184 DOI: 10.1177/0271678x221147366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Pericytes are the mural cells of the microvascular network that are in close contact with underlying endothelial cells. Endothelial-secreted PDGFB leads to recruitment of pericytes to the vessel wall, but this is disrupted in Pdgfbret/ret mice when the PDGFB retention motif is deleted. This results in severely reduced pericyte coverage on blood vessels. In this study, we investigated vascular abnormalities and hemodynamics in Pdgfbret/ret mice throughout the cerebrovascular network and in different cortical layers by in vivo two-photon microscopy. We confirmed that Pdgfbret/ret mice are severely deficient in pericytes throughout the vascular network, with enlarged brain blood vessels and a reduced number of vessel branches. Red blood cell velocity, linear density, and tube hematocrit were reduced in Pdgfbret/ret mice, which may impair oxygen delivery to the tissue. We also measured intravascular PO2 and found that concentrations were higher in cortical Layer 2/3 in Pdgfbret/ret mice, indicative of reduced blood oxygen extraction. Finally, we found that Pdgfbret/ret mice had a reduced capacity for vasodilation in response to an acetazolamide challenge during functional MRI imaging. Taken together, these results suggest that severe pericyte deficiency can lead to vascular abnormalities and altered cerebral blood flow, reminiscent of pathologies such as arteriovenous malformations.
Collapse
Affiliation(s)
- Jillian L Stobart
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.,Neuroscience Center, University and ETH Zurich, Zurich, Switzerland.,College of Pharmacy, University of Manitoba, Winnipeg, MB, Canada
| | - Eva Erlebach
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.,Neuroscience Center, University and ETH Zurich, Zurich, Switzerland
| | - Chaim Glück
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.,Neuroscience Center, University and ETH Zurich, Zurich, Switzerland
| | - Sheng-Fu Huang
- Neuroscience Center, University and ETH Zurich, Zurich, Switzerland.,Department of Neurosurgery, University Hospital Zurich, University of Zurich, Zürich, Switzerland
| | - Matthew Jp Barrett
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.,Neuroscience Center, University and ETH Zurich, Zurich, Switzerland
| | - Max Li
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.,Neuroscience Center, University and ETH Zurich, Zurich, Switzerland
| | - Sergei A Vinogradov
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jan Klohs
- Neuroscience Center, University and ETH Zurich, Zurich, Switzerland.,Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Yvette Zarb
- Neuroscience Center, University and ETH Zurich, Zurich, Switzerland.,Department of Neurosurgery, University Hospital Zurich, University of Zurich, Zürich, Switzerland
| | - Annika Keller
- Neuroscience Center, University and ETH Zurich, Zurich, Switzerland.,Department of Neurosurgery, University Hospital Zurich, University of Zurich, Zürich, Switzerland
| | - Bruno Weber
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.,Neuroscience Center, University and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
37
|
The Pathology of Primary Familial Brain Calcification: Implications for Treatment. Neurosci Bull 2022; 39:659-674. [PMID: 36469195 PMCID: PMC10073384 DOI: 10.1007/s12264-022-00980-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 07/10/2022] [Indexed: 12/08/2022] Open
Abstract
AbstractPrimary familial brain calcification (PFBC) is an inherited neurodegenerative disorder mainly characterized by progressive calcium deposition bilaterally in the brain, accompanied by various symptoms, such as dystonia, ataxia, parkinsonism, dementia, depression, headaches, and epilepsy. Currently, the etiology of PFBC is largely unknown, and no specific prevention or treatment is available. During the past 10 years, six causative genes (SLC20A2, PDGFRB, PDGFB, XPR1, MYORG, and JAM2) have been identified in PFBC. In this review, considering mechanistic studies of these genes at the cellular level and in animals, we summarize the pathogenesis and potential preventive and therapeutic strategies for PFBC patients. Our systematic analysis suggests a classification for PFBC genetic etiology based on several characteristics, provides a summary of the known composition of brain calcification, and identifies some potential therapeutic targets for PFBC.
Collapse
|
38
|
Wang X, Hui Q, Jin Z, Rao F, Jin L, Yu B, Banda J, Li X. Roles of growth factors in eye development and ophthalmic diseases. Zhejiang Da Xue Xue Bao Yi Xue Ban 2022; 51:613-625. [PMID: 36581579 PMCID: PMC10264994 DOI: 10.3724/zdxbyxb-2022-0603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 07/30/2022] [Indexed: 12/02/2022]
Abstract
Growth factors are active substances secreted by a variety of cells, which act as messengers to regulate cell migration, proliferation and differentiation. Many growth factors are involved in the eye development or the pathophysiological processes of eye diseases. Growth factors such as vascular endothelial growth factor and basic fibroblast growth factor mediate the occurrence and development of diabetic retinopathy, choroidal neovascularization, cataract, diabetic macular edema, and other retinal diseases. On the other hand, growth factors like nerve growth factor, ciliary neurotrophic factor, glial cell line-derived neurotrophic factor, pigment epithelial-derived factor and granulocyte colony-stimulating factor are known to promote optic nerve injury repair. Growth factors are also related to the pathogenesis of myopia. Fibroblast growth factor, transforming growth factor-β, and insulin-like growth factor regulate scleral thickness and influence the occurrence and development of myopia. This article reviews growth factors involved in ocular development and ocular pathophysiology, discusses the relationship between growth factors and ocular diseases, to provide reference for the application of growth factors in ophthalmology.
Collapse
|
39
|
Drapé E, Anquetil T, Larrivée B, Dubrac A. Brain arteriovenous malformation in hereditary hemorrhagic telangiectasia: Recent advances in cellular and molecular mechanisms. Front Hum Neurosci 2022; 16:1006115. [PMID: 36504622 PMCID: PMC9729275 DOI: 10.3389/fnhum.2022.1006115] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 10/27/2022] [Indexed: 11/25/2022] Open
Abstract
Hereditary hemorrhagic telangiectasia (HHT) is a genetic disorder characterized by vessel dilatation, such as telangiectasia in skin and mucosa and arteriovenous malformations (AVM) in internal organs such as the gastrointestinal tract, lungs, and brain. AVMs are fragile and tortuous vascular anomalies that directly connect arteries and veins, bypassing healthy capillaries. Mutations in transforming growth factor β (TGFβ) signaling pathway components, such as ENG (ENDOGLIN), ACVRL1 (ALK1), and SMAD4 (SMAD4) genes, account for most of HHT cases. 10-20% of HHT patients develop brain AVMs (bAVMs), which can lead to vessel wall rupture and intracranial hemorrhages. Though the main mutations are known, mechanisms leading to AVM formation are unclear, partially due to lack of animal models. Recent mouse models allowed significant advances in our understanding of AVMs. Endothelial-specific deletion of either Acvrl1, Eng or Smad4 is sufficient to induce AVMs, identifying endothelial cells (ECs) as primary targets of BMP signaling to promote vascular integrity. Loss of ALK1/ENG/SMAD4 signaling is associated with NOTCH signaling defects and abnormal arteriovenous EC differentiation. Moreover, cumulative evidence suggests that AVMs originate from venous ECs with defective flow-migration coupling and excessive proliferation. Mutant ECs show an increase of PI3K/AKT signaling and inhibitors of this signaling pathway rescue AVMs in HHT mouse models, revealing new therapeutic avenues. In this review, we will summarize recent advances and current knowledge of mechanisms controlling the pathogenesis of bAVMs, and discuss unresolved questions.
Collapse
Affiliation(s)
- Elise Drapé
- Centre de Recherche, CHU St. Justine, Montréal, QC, Canada,Département de Pharmacologie et de Physiologie, Université de Montréal, Montréal, QC, Canada
| | - Typhaine Anquetil
- Centre de Recherche, CHU St. Justine, Montréal, QC, Canada,Département De Pathologie et Biologie Cellulaire, Université de Montréal, Montréal, QC, Canada
| | - Bruno Larrivée
- Département d’Ophtalmologie, Université de Montréal, Montréal, QC, Canada,Centre De Recherche, Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada,*Correspondence: Bruno Larrivée,
| | - Alexandre Dubrac
- Centre de Recherche, CHU St. Justine, Montréal, QC, Canada,Département De Pathologie et Biologie Cellulaire, Université de Montréal, Montréal, QC, Canada,Département d’Ophtalmologie, Université de Montréal, Montréal, QC, Canada,Alexandre Dubrac,
| |
Collapse
|
40
|
Akasaka Y. The Role of Mesenchymal Stromal Cells in Tissue Repair and Fibrosis. Adv Wound Care (New Rochelle) 2022; 11:561-574. [PMID: 34841889 DOI: 10.1089/wound.2021.0037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Significance: The present review covers an overview of the current understanding of biology of mesenchymal stromal cells (MSCs) and suggests an important role of their differential potential for clinical approaches associated with tissue repair and fibrosis. Recent Advances: Genetic lineage tracing technology has enabled the delineation of cellular hierarchies and examination of MSC cellular origins and myofibroblast sources. This technique has led to the characterization of perivascular MSC populations and suggests that pericytes might provide a local source of tissue-specific MSCs, which can differentiate into tissue-specific cells for tissue repair and fibrosis. Autologous adipose tissue MSCs led to the advance in tissue engineering for regeneration of damaged tissues. Critical Issues: Recent investigation has revealed that perivascular MSCs might be the origin of myofibroblasts during fibrosis development, and perivascular MSCs might be the major source of myofibroblasts in fibrogenic disease. Adipose tissue MSCs combined with cytokines and biomaterials are available in the treatment of soft tissue defect and skin wound healing. Future Directions: Further investigation of the roles of perivascular MSCs may enable new approaches in the treatment of fibrogenic disease; moreover, perivascular MSCs might have potential as an antifibrotic target for fibrogenic disease. Autologous adipose tissue MSCs combined with cytokines and biomaterials will be an alternative method for the treatment of soft tissue defect and skin wound healing.
Collapse
Affiliation(s)
- Yoshikiyo Akasaka
- Division of Research Promotion and Development, Advanced Research Center, Toho University Graduate School of Medicine, Ota-ku, Japan.,Department of Pathology, Toho University School of Medicine, Ota-ku, Japan
| |
Collapse
|
41
|
Zhang Z, Warner KA, Mantesso A, Nör JE. PDGF-BB signaling via PDGFR-β regulates the maturation of blood vessels generated upon vasculogenic differentiation of dental pulp stem cells. Front Cell Dev Biol 2022; 10:977725. [PMID: 36340037 PMCID: PMC9627550 DOI: 10.3389/fcell.2022.977725] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 09/29/2022] [Indexed: 11/13/2022] Open
Abstract
A functional vascular network requires that blood vessels are invested by mural cells. We have shown that dental pulp stem cells (DPSC) can undergo vasculogenic differentiation, and that the resulting vessels anastomize with the host vasculature and become functional (blood carrying) vessels. However, the mechanisms underlying the maturation of DPSC-derived blood vessels remains unclear. Here, we performed a series of studies to understand the process of mural cell investment of blood vessels generated upon vasculogenic differentiation of dental pulp stem cells. Primary human DPSC were co-cultured with primary human umbilical artery smooth muscle cells (HUASMC) in 3D gels in presence of vasculogenic differentiation medium. We observed DPSC capillary sprout formation and SMC recruitment, alignment and remodeling that resulted in complex vascular networks. While HUASMC enhanced the number of capillary sprouts and stabilized the capillary network when co-cultured with DPSC, HUASMC by themselves were unable to form capillary sprouts. In vivo, GFP transduced human DPSC seeded in biodegradable scaffolds and transplanted into immunodeficient mice generated functional human blood vessels invested with murine smooth muscle actin (SMA)-positive, GFP-negative cells. Inhibition of PDGFR-β signaling prevented the SMC investment of DPSC-derived capillary sprouts in vitro and of DPSC-derived blood vessels in vivo. In contrast, inhibition of Tie-2 signaling did not have a significant effect on the SMC recruitment in DPSC-derived vascular structures. Collectively, these results demonstrate that PDGF-BB signaling via PDGFR-β regulates the process of maturation (mural investment) of blood vessels generated upon vasculogenic differentiation of human dental pulp stem cells.
Collapse
Affiliation(s)
- Zhaocheng Zhang
- Angiogenesis Research Laboratory, Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI, United States
| | - Kristy A. Warner
- Angiogenesis Research Laboratory, Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI, United States
| | - Andrea Mantesso
- Angiogenesis Research Laboratory, Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI, United States
| | - Jacques E. Nör
- Angiogenesis Research Laboratory, Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI, United States
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI, United States
- Department of Otolaryngology, University of Michigan School of Medicine, Ann Arbor, MI, United States
| |
Collapse
|
42
|
Amini AP, Kirkpatrick JD, Wang CS, Jaeger AM, Su S, Naranjo S, Zhong Q, Cabana CM, Jacks T, Bhatia SN. Multiscale profiling of protease activity in cancer. Nat Commun 2022; 13:5745. [PMID: 36192379 PMCID: PMC9530178 DOI: 10.1038/s41467-022-32988-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 08/24/2022] [Indexed: 11/09/2022] Open
Abstract
Diverse processes in cancer are mediated by enzymes, which most proximally exert their function through their activity. High-fidelity methods to profile enzyme activity are therefore critical to understanding and targeting the pathological roles of enzymes in cancer. Here, we present an integrated set of methods for measuring specific protease activities across scales, and deploy these methods to study treatment response in an autochthonous model of Alk-mutant lung cancer. We leverage multiplexed nanosensors and machine learning to analyze in vivo protease activity dynamics in lung cancer, identifying significant dysregulation that includes enhanced cleavage of a peptide, S1, which rapidly returns to healthy levels with targeted therapy. Through direct on-tissue localization of protease activity, we pinpoint S1 cleavage to the tumor vasculature. To link protease activity to cellular function, we design a high-throughput method to isolate and characterize proteolytically active cells, uncovering a pro-angiogenic phenotype in S1-cleaving cells. These methods provide a framework for functional, multiscale characterization of protease dysregulation in cancer.
Collapse
Affiliation(s)
- Ava P Amini
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Program in Biophysics, Harvard University, Boston, MA, USA
- Microsoft Research New England, Cambridge, MA, USA
| | - Jesse D Kirkpatrick
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Cathy S Wang
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Alex M Jaeger
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Susan Su
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Santiago Naranjo
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Qian Zhong
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Christina M Cabana
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Tyler Jacks
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sangeeta N Bhatia
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Harvard MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA.
- Wyss Institute at Harvard University, Boston, MA, USA.
- Howard Hughes Medical Institute, Cambridge, MA, USA.
| |
Collapse
|
43
|
Sharma K, Zhang Y, Paudel KR, Kachelmeier A, Hansbro PM, Shi X. The Emerging Role of Pericyte-Derived Extracellular Vesicles in Vascular and Neurological Health. Cells 2022; 11:cells11193108. [PMID: 36231071 PMCID: PMC9563036 DOI: 10.3390/cells11193108] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/27/2022] [Accepted: 09/30/2022] [Indexed: 12/02/2022] Open
Abstract
Pericytes (PCs), as a central component of the neurovascular unit, contribute to the regenerative potential of the central nervous system (CNS) and peripheral nervous system (PNS) by virtue of their role in blood flow regulation, angiogenesis, maintenance of the BBB, neurogenesis, and neuroprotection. Emerging evidence indicates that PCs also have a role in mediating cell-to-cell communication through the secretion of extracellular vesicles (EVs). Extracellular vesicles are cell-derived, micro- to nano-sized vesicles that transport cell constituents such as proteins, nucleic acids, and lipids from a parent originating cell to a recipient cell. PC-derived EVs (PC-EVs) play a crucial homeostatic role in neurovascular disease, as they promote angiogenesis, maintain the integrity of the blood-tissue barrier, and provide neuroprotection. The cargo carried by PC-EVs includes growth factors such as endothelial growth factor (VEGF), connecting tissue growth factors (CTGFs), fibroblast growth factors, angiopoietin 1, and neurotrophic growth factors such as brain-derived neurotrophic growth factor (BDNF), neuron growth factor (NGF), and glial-derived neurotrophic factor (GDNF), as well as cytokines such as interleukin (IL)-6, IL-8, IL-10, and MCP-1. The PC-EVs also carry miRNA and circular RNA linked to neurovascular health and the progression of several vascular and neuronal diseases. Therapeutic strategies employing PC-EVs have potential in the treatment of vascular and neurodegenerative diseases. This review discusses current research on the characteristic features of EVs secreted by PCs and their role in neuronal and vascular health and disease.
Collapse
Affiliation(s)
- Kushal Sharma
- Oregon Hearing Research Center, Department of Otolaryngology/Head & Neck Surgery, Oregon Health & Science University, Portland, OR 97239, USA
| | - Yunpei Zhang
- Oregon Hearing Research Center, Department of Otolaryngology/Head & Neck Surgery, Oregon Health & Science University, Portland, OR 97239, USA
| | - Keshav Raj Paudel
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW 2007, Australia
| | - Allan Kachelmeier
- Oregon Hearing Research Center, Department of Otolaryngology/Head & Neck Surgery, Oregon Health & Science University, Portland, OR 97239, USA
| | - Philip M. Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW 2007, Australia
| | - Xiaorui Shi
- Oregon Hearing Research Center, Department of Otolaryngology/Head & Neck Surgery, Oregon Health & Science University, Portland, OR 97239, USA
- Correspondence: ; Tel.: +1-503-494-2997
| |
Collapse
|
44
|
Neffeová K, Olejníčková V, Naňka O, Kolesová H. Development and diseases of the coronary microvasculature and its communication with the myocardium. WIREs Mech Dis 2022; 14:e1560. [DOI: 10.1002/wsbm.1560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 04/12/2022] [Accepted: 04/27/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Kristýna Neffeová
- Institute of Anatomy, First Faculty of Medicine Charles University Prague Czech Republic
| | - Veronika Olejníčková
- Institute of Anatomy, First Faculty of Medicine Charles University Prague Czech Republic
- Institute of Physiology Czech Academy of Science Prague Czech Republic
| | - Ondřej Naňka
- Institute of Anatomy, First Faculty of Medicine Charles University Prague Czech Republic
| | - Hana Kolesová
- Institute of Anatomy, First Faculty of Medicine Charles University Prague Czech Republic
- Institute of Physiology Czech Academy of Science Prague Czech Republic
| |
Collapse
|
45
|
Banerjee P, Rodning SP, Diniz WJS, Dyce PW. Co-Expression Network and Integrative Analysis of Metabolome and Transcriptome Uncovers Biological Pathways for Fertility in Beef Heifers. Metabolites 2022; 12:metabo12080708. [PMID: 36005579 PMCID: PMC9413342 DOI: 10.3390/metabo12080708] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 12/13/2022] Open
Abstract
Reproductive failure remains a significant challenge to the beef industry. The omics technologies have provided opportunities to improve reproductive efficiency. We used a multistaged analysis from blood profiles to integrate metabolome (plasma) and transcriptome (peripheral white blood cells) in beef heifers. We used untargeted metabolomics and RNA-Seq paired data from six AI-pregnant (AI-P) and six nonpregnant (NP) Angus-Simmental crossbred heifers at artificial insemination (AI). Based on network co-expression analysis, we identified 17 and 37 hub genes in the AI-P and NP groups, respectively. Further, we identified TGM2, TMEM51, TAC3, NDRG4, and PDGFB as more connected in the NP heifers’ network. The NP gene network showed a connectivity gain due to the rewiring of major regulators. The metabolomic analysis identified 18 and 15 hub metabolites in the AI-P and NP networks. Tryptophan and allantoic acid exhibited a connectivity gain in the NP and AI-P networks, respectively. The gene–metabolite integration identified tocopherol-a as positively correlated with ENSBTAG00000009943 in the AI-P group. Conversely, tocopherol-a was negatively correlated in the NP group with EXOSC2, TRNAUIAP, and SNX12. In the NP group, α-ketoglutarate-SMG8 and putrescine-HSD17B13 were positively correlated, whereas a-ketoglutarate-ALAS2 and tryptophan-MTMR1 were negatively correlated. These multiple interactions identified novel targets and pathways underlying fertility in bovines.
Collapse
|
46
|
Orlich MM, Diéguez-Hurtado R, Muehlfriedel R, Sothilingam V, Wolburg H, Oender CE, Woelffing P, Betsholtz C, Gaengel K, Seeliger M, Adams RH, Nordheim A. Mural Cell SRF Controls Pericyte Migration, Vessel Patterning and Blood Flow. Circ Res 2022; 131:308-327. [PMID: 35862101 PMCID: PMC9348820 DOI: 10.1161/circresaha.122.321109] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Pericytes and vascular smooth muscle cells, collectively known as mural cells, are recruited through PDGFB (platelet-derived growth factor B)-PDGFRB (platelet-derived growth factor receptor beta) signaling. MCs are essential for vascular integrity, and their loss has been associated with numerous diseases. Most of this knowledge is based on studies in which MCs are insufficiently recruited or fully absent upon inducible ablation. In contrast, little is known about the physiological consequences that result from impairment of specific MC functions. Here, we characterize the role of the transcription factor SRF (serum response factor) in MCs and study its function in developmental and pathological contexts.
Collapse
Affiliation(s)
- Michael M. Orlich
- Department of Molecular Biology, Interfaculty Institute for Cell Biology, University of Tuebingen, Germany (M.M.O., C.E.O., P.W., A.N.)
- International Max Planck Research School (IMPRS) “From Molecules to Organisms,” Tuebingen, Germany (M.M.O., A.N.)
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (M.M.O., C.B., K.G.)
- Now with Rudbeck Laboratory C11, Dag Hammarskjölds Väg 20, 751 85 Uppsala, Sweden (M.M.O.)
| | - Rodrigo Diéguez-Hurtado
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Muenster, Germany (R.D.-H., R.H.A.)
- Faculty of Medicine, University of Muenster, Muenster, Germany (R.D.-H., R.H.A.)
| | - Regine Muehlfriedel
- Institute for Ophthalmic Research, Centre for Ophthalmology, University Clinic Tuebingen (UKT), Germany. (R.M., V.S., M.S.)
| | - Vithiyanjali Sothilingam
- Institute for Ophthalmic Research, Centre for Ophthalmology, University Clinic Tuebingen (UKT), Germany. (R.M., V.S., M.S.)
| | - Hartwig Wolburg
- Department of General Pathology and Pathological Anatomy, Institute of Pathology and Neuropathology, University Clinic Tuebingen (UKT), Germany. (H.W.)
| | - Cansu Ebru Oender
- Department of Molecular Biology, Interfaculty Institute for Cell Biology, University of Tuebingen, Germany (M.M.O., C.E.O., P.W., A.N.)
| | - Pascal Woelffing
- Department of Molecular Biology, Interfaculty Institute for Cell Biology, University of Tuebingen, Germany (M.M.O., C.E.O., P.W., A.N.)
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (M.M.O., C.B., K.G.)
| | - Konstantin Gaengel
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (M.M.O., C.B., K.G.)
| | - Mathias Seeliger
- Institute for Ophthalmic Research, Centre for Ophthalmology, University Clinic Tuebingen (UKT), Germany. (R.M., V.S., M.S.)
| | - Ralf H. Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Muenster, Germany (R.D.-H., R.H.A.)
- Faculty of Medicine, University of Muenster, Muenster, Germany (R.D.-H., R.H.A.)
| | - Alfred Nordheim
- Department of Molecular Biology, Interfaculty Institute for Cell Biology, University of Tuebingen, Germany (M.M.O., C.E.O., P.W., A.N.)
- International Max Planck Research School (IMPRS) “From Molecules to Organisms,” Tuebingen, Germany (M.M.O., A.N.)
| |
Collapse
|
47
|
Ben-Zvi A, Liebner S. Developmental regulation of barrier- and non-barrier blood vessels in the CNS. J Intern Med 2022; 292:31-46. [PMID: 33665890 DOI: 10.1111/joim.13263] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 02/01/2021] [Indexed: 12/22/2022]
Abstract
The blood-brain barrier (BBB) is essential for creating and maintaining tissue homeostasis in the central nervous system (CNS), which is key for proper neuronal function. In most vertebrates, the BBB is localized to microvascular endothelial cells that acquire barrier properties during angiogenesis of the neuroectoderm. Complex and continuous tight junctions, and the lack of fenestrae combined with low pinocytotic activity render the BBB endothelium a tight barrier for water-soluble molecules that may only enter the CNS via specific transporters. The differentiation of these unique endothelial properties during embryonic development is initiated by endothelial-specific flavours of the Wnt/β-catenin pathway in a precise spatiotemporal manner. In this review, we summarize the currently known cellular (neural precursor and endothelial cells) and molecular (VEGF and Wnt/β-catenin) mechanisms mediating brain angiogenesis and barrier formation. Moreover, we introduce more recently discovered crosstalk with cellular and acellular elements within the developing CNS such as the extracellular matrix. We discuss recent insights into the downstream molecular mechanisms of Wnt/β-catenin in particular, the recently identified target genes like Foxf2, Foxl2, Foxq1, Lef1, Ppard, Zfp551, Zic3, Sox17, Apcdd1 and Fgfbp1 that are involved in refining and maintaining barrier characteristics in the mature BBB endothelium. Additionally, we elute to recent insight into barrier heterogeneity and differential endothelial barrier properties within the CNS, focussing on the circumventricular organs as well as on the neurogenic niches in the subventricular zone and the hippocampus. Finally, open questions and future BBB research directions are highlighted in the context of taking benefit from understanding BBB development for strategies to modulate BBB function under pathological conditions.
Collapse
Affiliation(s)
- A Ben-Zvi
- From the, The Department of Developmental Biology and Cancer Research, Institute for Medical Research IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - S Liebner
- Institute of Neurology (Edinger Institute), University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany.,Excellence Cluster Cardio-Pulmonary Systems (ECCPS), Partner Site Frankfurt, Frankfurt am Main, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Frankfurt am Main, Germany
| |
Collapse
|
48
|
Matsuoka RL, Buck LD, Vajrala KP, Quick RE, Card OA. Historical and current perspectives on blood endothelial cell heterogeneity in the brain. Cell Mol Life Sci 2022; 79:372. [PMID: 35726097 PMCID: PMC9209386 DOI: 10.1007/s00018-022-04403-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/18/2022] [Accepted: 05/25/2022] [Indexed: 11/28/2022]
Abstract
Dynamic brain activity requires timely communications between the brain parenchyma and circulating blood. Brain-blood communication is facilitated by intricate networks of brain vasculature, which display striking heterogeneity in structure and function. This vascular cell heterogeneity in the brain is fundamental to mediating diverse brain functions and has long been recognized. However, the molecular basis of this biological phenomenon has only recently begun to be elucidated. Over the past century, various animal species and in vitro systems have contributed to the accumulation of our fundamental and phylogenetic knowledge about brain vasculature, collectively advancing this research field. Historically, dye tracer and microscopic observations have provided valuable insights into the anatomical and functional properties of vasculature across the brain, and these techniques remain an important approach. Additionally, recent advances in molecular genetics and omics technologies have revealed significant molecular heterogeneity within brain endothelial and perivascular cell types. The combination of these conventional and modern approaches has enabled us to identify phenotypic differences between healthy and abnormal conditions at the single-cell level. Accordingly, our understanding of brain vascular cell states during physiological, pathological, and aging processes has rapidly expanded. In this review, we summarize major historical advances and current knowledge on blood endothelial cell heterogeneity in the brain, and discuss important unsolved questions in the field.
Collapse
Affiliation(s)
- Ryota L Matsuoka
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA. .,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA.
| | - Luke D Buck
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA
| | - Keerti P Vajrala
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA.,Kansas City University College of Osteopathic Medicine, Kansas City, MO 64106, USA
| | - Rachael E Quick
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA
| | - Olivia A Card
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA
| |
Collapse
|
49
|
Nielson CD, Berthiaume AA, Bonney SK, Shih AY. In vivo Single Cell Optical Ablation of Brain Pericytes. Front Neurosci 2022; 16:900761. [PMID: 35720702 PMCID: PMC9205398 DOI: 10.3389/fnins.2022.900761] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/19/2022] [Indexed: 01/09/2023] Open
Abstract
Pericytes have myriad functions in cerebrovascular regulation but remain understudied in the living brain. To dissect pericyte functions in vivo, prior studies have used genetic approaches to induce global pericyte loss in the rodent brain. However, this leads to complex outcomes, making it challenging to disentangle the physiological roles of pericytes from the pathophysiological effects of their depletion. Here, we describe a protocol to optically ablate individual pericytes of the mouse cerebral cortex in vivo for fine-scale studies of pericyte function. The strategy relies on two-photon microscopy and cranial window-implanted transgenic mice with mural cell-specific expression of fluorescent proteins. Single pericyte somata are precisely targeted with pulsed infrared laser light to induce selective pericyte death, but without overt blood-brain barrier leakage. Following pericyte ablation, the changes to the local capillary network and remaining pericytes can be examined longitudinally. The approach has been used to study pericyte roles in capillary flow regulation, and the structural remodeling of pericytes involved in restoration of endothelial coverage after pericyte loss.
Collapse
Affiliation(s)
- Cara D. Nielson
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA, United States,Graduate Program in Neuroscience, University of Washington, Seattle, WA, United States
| | - Andrée-Anne Berthiaume
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA, United States,Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - Stephanie K. Bonney
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Andy Y. Shih
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA, United States,Department of Pediatrics, University of Washington, Seattle, WA, United States,Department of Bioengineering, University of Washington, Seattle, WA, United States,*Correspondence: Andy Y. Shih,
| |
Collapse
|
50
|
Manocha E, Consonni A, Baggi F, Ciusani E, Cocce V, Paino F, Tremolada C, Caruso A, Alessandri G. CD146+ Pericytes Subset Isolated from Human Micro-Fragmented Fat Tissue Display a Strong Interaction with Endothelial Cells: A Potential Cell Target for Therapeutic Angiogenesis. Int J Mol Sci 2022; 23:ijms23105806. [PMID: 35628617 PMCID: PMC9144360 DOI: 10.3390/ijms23105806] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 02/04/2023] Open
Abstract
Pericytes (PCs) are mesenchymal stromal cells (MSCs) that function as support cells and play a role in tissue regeneration and, in particular, vascular homeostasis. PCs promote endothelial cells (ECs) survival which is critical for vessel stabilization, maturation, and remodeling. In this study, PCs were isolated from human micro-fragmented adipose tissue (MFAT) obtained from fat lipoaspirate and were characterized as NG2+/PDGFRβ+/CD105+ cells. Here, we tested the fat-derived PCs for the dispensability of the CD146 marker with the aim of better understanding the role of these PC subpopulations on angiogenesis. Cells from both CD146-positive (CD146+) and negative (CD146−) populations were observed to interact with human umbilical vein ECs (HUVECs). In addition, fat-derived PCs were able to induce angiogenesis of ECs in spheroids assay; and conditioned medium (CM) from both PCs and fat tissue itself led to the proliferation of ECs, thereby marking their role in angiogenesis stimulation. However, we found that CD146+ cells were more responsive to PDGF-BB-stimulated migration, adhesion, and angiogenic interaction with ECs, possibly owing to their higher expression of NCAM/CD56 than the corresponding CD146− subpopulation. We conclude that in fat tissue, CD146-expressing cells may represent a more mature pericyte subpopulation that may have higher efficacy in controlling and stimulating vascular regeneration and stabilization than their CD146-negative counterpart.
Collapse
Affiliation(s)
- Ekta Manocha
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia Medical School, 25123 Brescia, Italy; (A.C.); (G.A.)
- Correspondence:
| | - Alessandra Consonni
- Neurology IV—Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (A.C.); (F.B.)
| | - Fulvio Baggi
- Neurology IV—Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (A.C.); (F.B.)
| | - Emilio Ciusani
- Laboratory of Neurological Biochemistry and Neuropharmacology, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy;
| | - Valentina Cocce
- CRC StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy; (V.C.); (F.P.)
| | - Francesca Paino
- CRC StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy; (V.C.); (F.P.)
| | - Carlo Tremolada
- Department of Stem Cells and Regenerative Medicine, Image Institute, 20122 Milan, Italy;
| | - Arnaldo Caruso
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia Medical School, 25123 Brescia, Italy; (A.C.); (G.A.)
| | - Giulio Alessandri
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia Medical School, 25123 Brescia, Italy; (A.C.); (G.A.)
- Department of Stem Cells and Regenerative Medicine, Image Institute, 20122 Milan, Italy;
| |
Collapse
|