1
|
Waxman S, Villafranca-Baughman D, Phillippi J, Jakobs TC, Alarcon-Martinez L, Di Polo A, Sigal IA. Pericytes in the Optic Nerve Head. Prog Retin Eye Res 2025:101375. [PMID: 40449651 DOI: 10.1016/j.preteyeres.2025.101375] [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: 01/04/2025] [Revised: 05/28/2025] [Accepted: 05/29/2025] [Indexed: 06/03/2025]
Abstract
Pericytes are a unique population of contractile mural cells and an essential part of the microvasculature. In the retina and brain, pericytes play crucial roles in regulating blood flow, maintaining the blood-brain barrier, signaling with neighboring cells, and depositing extracellular matrix. Pericyte dysfunction is an early process in a variety of neurodegenerative conditions. However, remarkably little is known about pericytes at an early site of neurodegeneration in glaucoma, the optic nerve head (ONH). This work summarizes the current understanding of pericyte contributions to ONH physiology, identifies potential roles in glaucomatous pathophysiology, and uncovers open questions at the intersection of these areas. We surveyed the literature to identify the roles of ONH pericytes in the context of health and glaucoma. Additionally, we probed for the presence of pericytes along microvasculature in mouse, nonhuman primate, and human donor ONH tissues. We identified an association between factors influencing ONH dysfunction in glaucoma and factors influencing pericyte dysfunction in other neurodegenerative conditions. Pericytes exist in the mouse, nonhuman primate, and human ONH, implicating their capacity for local function. ONH pericytes represent a promising but underexplored target for treating microvascular impairment in glaucoma. Investigating the contribution of pericytes in both healthy and disease states can help inform mechanisms of dysfunction in glaucomatous pathology, paving the way for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Susannah Waxman
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh PA, USA
| | - Deborah Villafranca-Baughman
- Department of Neuroscience, Université de Montréal, Montréal, QC; Canada Neuroscience Division, Centre de Recherche du Centre Hospitalier, Université de Montréal, Montréal, QC, Canada
| | - Julie Phillippi
- Division of Cardiac Surgery, Department of Cardiothoracic Surgery, University of Pittsburgh
| | - Tatjana C Jakobs
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary/Schepens Eye Research Institute, Harvard Medical School, Boston, MA, USA
| | - Luis Alarcon-Martinez
- Centre for Eye Research Australia, Department of Ophthalmology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Adriana Di Polo
- Department of Neuroscience, Université de Montréal, Montréal, QC; Canada Neuroscience Division, Centre de Recherche du Centre Hospitalier, Université de Montréal, Montréal, QC, Canada
| | - Ian A Sigal
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh PA, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh PA, USA.
| |
Collapse
|
2
|
Meza-Resillas J, O'Hara F, Kaushik S, Stobart MJ, Ahmadpour N, Kantroo M, Del Rosario J, Rodriguez MC, Koval D, Glück C, Weber B, Stobart JL. Systemic nimodipine affects pericyte calcium signaling, resting hemodynamics and neurovascular coupling in healthy mouse brain. Neurotherapeutics 2025:e00614. [PMID: 40404520 DOI: 10.1016/j.neurot.2025.e00614] [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: 12/01/2024] [Revised: 04/28/2025] [Accepted: 05/13/2025] [Indexed: 05/24/2025] Open
Abstract
Nimodipine is a L-type voltage gated calcium channel blocker commonly given to patients after a sub-arachnoid hemorrhage. It is known to dilate cerebral arteries and affect brain pericytes that express voltage gated calcium channels. Here, we systemically administered nimodipine (1 mg/kg; i.p.) and measured brain pericyte calcium transients and single-vessel hemodynamics in the brains of mice by two-photon microscopy. We find that nimodipine reduces calcium transients in all types of pericytes, from ensheathing to thin-strand cells, at different locations in the vascular network. This induces local vasodilation of vessels closer to penetrating arterioles but decreases blood cell velocity. These vascular consequences are due to systemic nimodipine effects because direct brain application of nimodipine caused blood cell velocity to increase. Nimodipine treatment also reduced further dilation during neurovascular coupling throughout the vascular network. Overall, this suggests that nimodipine can change cerebrovascular hemodynamics by altering pericyte physiology and these are important considerations for the clinical use of this drug.
Collapse
Affiliation(s)
- Jessica Meza-Resillas
- College of Pharmacy, University of Manitoba, Winnipeg, Manitoba, Canada; Centre on Aging, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Finnegan O'Hara
- College of Pharmacy, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Syed Kaushik
- College of Pharmacy, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Michael J Stobart
- College of Pharmacy, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Noushin Ahmadpour
- College of Pharmacy, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Meher Kantroo
- College of Pharmacy, University of Manitoba, Winnipeg, Manitoba, Canada
| | - John Del Rosario
- College of Pharmacy, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Megan C Rodriguez
- College of Pharmacy, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Dmytro Koval
- College of Pharmacy, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Chaim Glück
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Bruno Weber
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Jillian L Stobart
- College of Pharmacy, University of Manitoba, Winnipeg, Manitoba, Canada; Centre on Aging, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
3
|
Dalkara T, Østergaard L, Heusch G, Attwell D. Pericytes in the brain and heart: functional roles and response to ischaemia and reperfusion. Cardiovasc Res 2025; 120:2336-2348. [PMID: 39074200 PMCID: PMC11976724 DOI: 10.1093/cvr/cvae147] [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: 02/05/2024] [Revised: 03/28/2024] [Accepted: 05/03/2024] [Indexed: 07/31/2024] Open
Abstract
In the last 20 years, there has been a revolution in our understanding of how blood flow is regulated in many tissues. Whereas it used to be thought that essentially all blood flow control occurred at the arteriole level, it is now recognized that control of capillary blood flow by contractile pericytes plays a key role both in regulating blood flow physiologically and in reducing it in clinically relevant pathological conditions. In this article, we compare and contrast how brain and cardiac pericytes regulate cerebral and coronary blood flow, focusing mainly on the pathological events of cerebral and cardiac ischaemia. The cerebral and coronary capillary beds differ dramatically in morphology, yet in both cases, pericyte-mediated capillary constriction plays a key role in restricting blood flow after ischaemia and possibly in other pathological conditions. We conclude with suggestions for therapeutic approaches to relaxing pericytes, which may prove useful in the long-term for reducing pericyte-induced ischaemia.
Collapse
Affiliation(s)
- Turgay Dalkara
- Department of Neuroscience, Bilkent University, Ankara 06800 Türkiye
- Department of Molecular Biology and Genetics, Bilkent University, Ankara 06800 Türkiye
| | - Leif Østergaard
- Center of Functionally Integrative Neuroscience (CFIN), Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Duisburg-Essen, Essen, Germany
| | - David Attwell
- Department of Neuroscience, Physiology & Pharmacology, University College London, Gower St., London WC1E 6BT, UK
| |
Collapse
|
4
|
Bakker N, Croes AA, Prevaes E, van Noorden CJF, Schlingemann RO, Klaassen I. Development of Immunostaining Protocols for 3D Visualization of Pericytes in Human Retinal Flatmounts. J Histochem Cytochem 2025; 73:147-170. [PMID: 40098221 PMCID: PMC11915233 DOI: 10.1369/00221554251323655] [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: 12/29/2024] [Accepted: 02/11/2025] [Indexed: 03/19/2025] Open
Abstract
Vascular pericytes are widely present across the human body and crucial in regulating vascular flow, permeability, and homeostasis. In the human retina, pericytes are important for forming and maintaining the blood-retinal barrier, as well as for autoregulation of blood flow. Pericyte loss has been implicated in various pathological conditions. Visualization of pericytes by immunofluorescence (IF) staining provides valuable information on pericyte number, morphology, location, and on expression of anatomic and functional markers. However, species-specific differences in pericyte marker expression exist. In this study, we aimed to develop a novel IF co-staining protocol to detect the pericyte markers NG2, PDGFRβ, αSMA, CD13, and RFC1 in human retinal flatmounts. Unlike retinal sections, retinal flatmounts enable 3D visualization of pericyte distribution across the entire vascular network. Key optimizations included tailoring the fixation method, blocking buffer composition and antibody solvent, as well as using jasplakinolide to enhance αSMA detection. Our protocol successfully enabled double staining of NG2 and PDGFRβ, as well as αSMA and PDGFRβ, whereas CD13 and RFC1 expression was not detectable in human retinal flatmounts. This novel 3D IF protocol enhances in situ visualization of human retinal pericytes, enabling accurate studies of their role in vascular health and disease to aid targeted therapy development.
Collapse
Affiliation(s)
- Noëlle Bakker
- Ocular Angiogenesis Group, Department of Ophthalmology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Amsterdam, The Netherlands
| | - Aïcha A. Croes
- Ocular Angiogenesis Group, Department of Ophthalmology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Eva Prevaes
- Ocular Angiogenesis Group, Department of Ophthalmology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Cornelis J. F. van Noorden
- Ocular Angiogenesis Group, Department of Ophthalmology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Reinier O. Schlingemann
- Ocular Angiogenesis Group, Department of Ophthalmology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Amsterdam, The Netherlands
- Department of Ophthalmology, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, University of Lausanne, Lausanne, Switzerland (ROS)
| | - Ingeborg Klaassen
- Ocular Angiogenesis Group, Department of Ophthalmology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Amsterdam, The Netherlands
| |
Collapse
|
5
|
Shiga Y, Rangel Olguin AG, El Hajji S, Belforte N, Quintero H, Dotigny F, Alarcon-Martinez L, Krishnaswamy A, Di Polo A. Endoplasmic reticulum stress-related deficits in calcium clearance promote neuronal dysfunction that is prevented by SERCA2 gene augmentation. Cell Rep Med 2024; 5:101839. [PMID: 39615485 PMCID: PMC11722116 DOI: 10.1016/j.xcrm.2024.101839] [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: 02/26/2024] [Revised: 07/25/2024] [Accepted: 11/04/2024] [Indexed: 12/20/2024]
Abstract
Disruption of calcium (Ca2+) homeostasis in neurons is a hallmark of neurodegenerative diseases. Here, we investigate the mechanisms leading to Ca2+ dysregulation and ask whether altered Ca2+ dynamics impinge on neuronal stress and circuit dysfunction. Using two-photon microscopy, we show that ocular hypertension, a major risk factor in glaucoma, and optic nerve crush injury disrupt the capacity of retinal neurons to clear cytosolic Ca2+ leading to impaired light-evoked responses. Gene- and protein expression analysis reveal the loss of the sarco-endoplasmic reticulum (ER) Ca2+-ATPase2 pump (SERCA2/ATP2A2) in injured retinal neurons from mice and patients with primary open-angle glaucoma. Pharmacological activation or neuron-specific gene delivery of SERCA2 is sufficient to rescue single-cell Ca2+ dynamics and promote robust survival of damaged neurons. Furthermore, SERCA2 gene supplementation reduces ER stress, reestablishes circuit balance, and restores visual behaviors. Our findings reveal that enhancing the Ca2+ clearance capacity of vulnerable neurons alleviates organelle stress and promotes neurorecovery.
Collapse
Affiliation(s)
- Yukihiro Shiga
- Department of Neuroscience, University of Montreal, PO box 6128, Station Centre-ville, Montreal, Quebec H3C 3J7, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, Quebec H2X 0A9, Canada
| | | | - Sana El Hajji
- Department of Neuroscience, University of Montreal, PO box 6128, Station Centre-ville, Montreal, Quebec H3C 3J7, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, Quebec H2X 0A9, Canada
| | - Nicolas Belforte
- Department of Neuroscience, University of Montreal, PO box 6128, Station Centre-ville, Montreal, Quebec H3C 3J7, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, Quebec H2X 0A9, Canada
| | - Heberto Quintero
- Department of Neuroscience, University of Montreal, PO box 6128, Station Centre-ville, Montreal, Quebec H3C 3J7, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, Quebec H2X 0A9, Canada
| | - Florence Dotigny
- Department of Neuroscience, University of Montreal, PO box 6128, Station Centre-ville, Montreal, Quebec H3C 3J7, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, Quebec H2X 0A9, Canada
| | - Luis Alarcon-Martinez
- Department of Neuroscience, University of Montreal, PO box 6128, Station Centre-ville, Montreal, Quebec H3C 3J7, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, Quebec H2X 0A9, Canada
| | - Arjun Krishnaswamy
- Department of Physiology, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Adriana Di Polo
- Department of Neuroscience, University of Montreal, PO box 6128, Station Centre-ville, Montreal, Quebec H3C 3J7, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, Quebec H2X 0A9, Canada.
| |
Collapse
|
6
|
Bohler F, Bohler L, Taranikanti V. Targeting pericyte retention in Diabetic Retinopathy: a review. Ann Med 2024; 56:2398200. [PMID: 39268600 PMCID: PMC11404372 DOI: 10.1080/07853890.2024.2398200] [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: 02/10/2024] [Revised: 07/26/2024] [Accepted: 08/13/2024] [Indexed: 09/17/2024] Open
Abstract
Diabetic retinopathy is a common yet severe complication of diabetes mellitus and is the leading cause of blindness in middle-aged adults. After years of poorly managed hyperglycemia, complications begin as non-proliferative diabetic retinopathy but can then progress into the proliferative stage marked by neovascularization of the retina. Multiple pathologic mechanisms caused by chronic hyperglycemia damage the retinal vasculature leading to pericyte drop out and the progression of the disease. This review outlines the major pathways of pathogenesis in diabetic retinopathy, highlighting the protective role pericytes play in preserving the blood-retinal barrier. Given the loss of this cell line is a defining feature of the disease, ways in which to prevent pericyte dropout within retinal vasculature is discussed, targeting various pathogenesis pathways of diabetic retinopathy.
Collapse
Affiliation(s)
- Forrest Bohler
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI
| | - Lily Bohler
- College of Letters and Science, Montana State University, Bozeman, MT
| | - Varna Taranikanti
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI
| |
Collapse
|
7
|
Raj S, Sarangi P, Goyal D, Kumar H. The Hidden Hand in White Matter: Pericytes and the Puzzle of Demyelination. ACS Pharmacol Transl Sci 2024; 7:2912-2923. [PMID: 39421660 PMCID: PMC11480894 DOI: 10.1021/acsptsci.4c00192] [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: 04/03/2024] [Revised: 08/01/2024] [Accepted: 08/27/2024] [Indexed: 10/19/2024]
Abstract
Disruption of myelin, the fatty sheath-insulating nerve fibers in the white matter, blocks or slows the rapid transmission of electrical signals along nerve cells and contributes to several neurodegenerative diseases such as multiple sclerosis. Traditionally, research has focused on neuronal dysfunction as the primary factor, including autoimmunity, infections, inflammation, and genetic disorders causing demyelination. However, recent insights emphasize the critical role of pericytes, non-neuronal cells that regulate blood flow and maintain the health of blood vessels within white matter. This Perspective explores the principal mechanisms through which pericyte dysfunction contributes to damage and demyelination, including impaired communication with neurons (neurovascular uncoupling), excessive formation of scar tissue (fibrosis), and the infiltration of detrimental substances from the bloodstream. Understanding these mechanisms of pericyte-driven demyelination may lead to the creation of new therapeutic strategies for tackling a range of neurodegenerative conditions.
Collapse
Affiliation(s)
- Siddharth Raj
- Department of Pharmacology
and Toxicology, National Institute of Pharmaceutical
Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India, 382355
| | - Priyabrata Sarangi
- Department of Pharmacology
and Toxicology, National Institute of Pharmaceutical
Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India, 382355
| | - Divya Goyal
- Department of Pharmacology
and Toxicology, National Institute of Pharmaceutical
Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India, 382355
| | - Hemant Kumar
- Department of Pharmacology
and Toxicology, National Institute of Pharmaceutical
Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India, 382355
| |
Collapse
|
8
|
Zhang S, Chen Y, Chen Q, Chen H, Wei L, Wang S. Assessment of cerebrovascular alterations induced by inflammatory response and oxidative-nitrative stress after traumatic intracranial hypertension and a potential mitigation strategy. Sci Rep 2024; 14:14535. [PMID: 38914585 PMCID: PMC11196732 DOI: 10.1038/s41598-024-64940-6] [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: 03/14/2024] [Accepted: 06/14/2024] [Indexed: 06/26/2024] Open
Abstract
The rapid perfusion of cerebral arteries leads to a significant increase in intracranial blood volume, exposing patients with traumatic brain injury to the risk of diffuse brain swelling or malignant brain herniation during decompressive craniectomy. The microcirculation and venous system are also involved in this process, but the precise mechanisms remain unclear. A physiological model of extremely high intracranial pressure was created in rats. This development triggered the TNF-α/NF-κB/iNOS axis in microglia, and released many inflammatory factors and reactive oxygen species/reactive nitrogen species, generating an excessive amount of peroxynitrite. Subsequently, the capillary wall cells especially pericytes exhibited severe degeneration and injury, the blood-brain barrier was disrupted, and a large number of blood cells were deposited within the microcirculation, resulting in a significant delay in the recovery of the microcirculation and venous blood flow compared to arterial flow, and this still persisted after decompressive craniectomy. Infliximab is a monoclonal antibody bound to TNF-α that effectively reduces the activity of TNF-α/NF-κB/iNOS axis. Treatment with Infliximab resulted in downregulation of inflammatory and oxidative-nitrative stress related factors, attenuation of capillary wall cells injury, and relative reduction of capillary hemostasis. These improved the delay in recovery of microcirculation and venous blood flow.
Collapse
Affiliation(s)
- Shangming Zhang
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, 350025, China
- Department of Neurosurgery, 900th Hospital, Fujian Provincial Clinical Medical Research Center for Minimally Invasive Diagnosis and Treatment of Neurovascular Diseases, Fuzhou, 350025, China
| | - Yehuang Chen
- Department of Neurosurgery, 900th Hospital, Fujian Provincial Clinical Medical Research Center for Minimally Invasive Diagnosis and Treatment of Neurovascular Diseases, Fuzhou, 350025, China
| | - Qizuan Chen
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, 350025, China
- Department of Neurosurgery, 900th Hospital, Fujian Provincial Clinical Medical Research Center for Minimally Invasive Diagnosis and Treatment of Neurovascular Diseases, Fuzhou, 350025, China
| | - Hongjie Chen
- Department of Neurosurgery, 900th Hospital, Fujian Provincial Clinical Medical Research Center for Minimally Invasive Diagnosis and Treatment of Neurovascular Diseases, Fuzhou, 350025, China
| | - Liangfeng Wei
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, 350025, China.
- Department of Neurosurgery, 900th Hospital, Fujian Provincial Clinical Medical Research Center for Minimally Invasive Diagnosis and Treatment of Neurovascular Diseases, Fuzhou, 350025, China.
| | - Shousen Wang
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, 350025, China.
- Department of Neurosurgery, 900th Hospital, Fujian Provincial Clinical Medical Research Center for Minimally Invasive Diagnosis and Treatment of Neurovascular Diseases, Fuzhou, 350025, China.
| |
Collapse
|
9
|
Sun F, Zhou J, Chen X, Yang T, Wang G, Ge J, Zhang Z, Mei Z. No-reflow after recanalization in ischemic stroke: From pathomechanisms to therapeutic strategies. J Cereb Blood Flow Metab 2024; 44:857-880. [PMID: 38420850 PMCID: PMC11318407 DOI: 10.1177/0271678x241237159] [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: 02/10/2023] [Revised: 01/07/2024] [Accepted: 02/18/2024] [Indexed: 03/02/2024]
Abstract
Endovascular reperfusion therapy is the primary strategy for acute ischemic stroke. No-reflow is a common phenomenon, which is defined as the failure of microcirculatory reperfusion despite clot removal by thrombolysis or mechanical embolization. It has been reported that up to 25% of ischemic strokes suffer from no-reflow, which strongly contributes to an increased risk of poor clinical outcomes. No-reflow is associated with functional and structural alterations of cerebrovascular microcirculation, and the injury to the microcirculation seriously hinders the neural functional recovery following macrovascular reperfusion. Accumulated evidence indicates that pathology of no-reflow is linked to adhesion, aggregation, and rolling of blood components along the endothelium, capillary stagnation with neutrophils, astrocytes end-feet, and endothelial cell edema, pericyte contraction, and vasoconstriction. Prevention or treatment strategies aim to alleviate or reverse these pathological changes, including targeted therapies such as cilostazol, adhesion molecule blocking antibodies, peroxisome proliferator-activated receptors (PPARs) activator, adenosine, pericyte regulators, as well as adjunctive therapies, such as extracorporeal counterpulsation, ischemic preconditioning, and alternative or complementary therapies. Herein, we provide an overview of pathomechanisms, predictive factors, diagnosis, and intervention strategies for no-reflow, and attempt to convey a new perspective on the clinical management of no-reflow post-ischemic stroke.
Collapse
Affiliation(s)
- Feiyue Sun
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jing Zhou
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xiangyu Chen
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Tong Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Guozuo Wang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jinwen Ge
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan Academy of Chinese Medicine, Changsha, Hunan, China
| | - Zhanwei Zhang
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Zhigang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, College of Medicine and Health Sciences, China Three Gorges University, Yichang, Hubei, China
| |
Collapse
|
10
|
Zhang Q, Yan X, Han H, Wang Y, Sun J. Pericyte in retinal vascular diseases: A multifunctional regulator and potential therapeutic target. FASEB J 2024; 38:e23679. [PMID: 38780117 DOI: 10.1096/fj.202302624r] [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: 12/19/2023] [Revised: 04/17/2024] [Accepted: 05/07/2024] [Indexed: 05/25/2024]
Abstract
Retinal vascular diseases (RVDs), in particular diabetic retinopathy, retinal vein occlusion, and retinopathy of prematurity, are leading contributors to blindness. The pathogenesis of RVD involves vessel dilatation, leakage, and occlusion; however, the specific underlying mechanisms remain unclear. Recent findings have indicated that pericytes (PCs), as critical members of the vascular mural cells, significantly contribute to the progression of RVDs, including detachment from microvessels, alteration of contractile and secretory properties, and excessive production of the extracellular matrix. Moreover, PCs are believed to have mesenchymal stem properties and, therefore, might contribute to regenerative therapy. Here, we review novel ideas concerning PC characteristics and functions in RVDs and discuss potential therapeutic strategies based on PCs, including the targeting of pathological signals and cell-based regenerative treatments.
Collapse
Affiliation(s)
- Quan Zhang
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Air Force Medical University, Xi'an, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Air Force Medical University, Xi'an, China
| | - Xianchun Yan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Air Force Medical University, Xi'an, China
| | - Hua Han
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Air Force Medical University, Xi'an, China
| | - Yusheng Wang
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Jiaxing Sun
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Air Force Medical University, Xi'an, China
- Department of Neurobiology, Air Force Medical University, Xi'an, China
| |
Collapse
|
11
|
Lu B, Li Y, Xie L, Chiu K, Hao X, Xu J, Luo J, Sham PC. Computational Retinal Microvascular Biomarkers from an OCTA Image in Clinical Investigation. Biomedicines 2024; 12:868. [PMID: 38672222 PMCID: PMC11048516 DOI: 10.3390/biomedicines12040868] [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: 12/25/2023] [Revised: 03/24/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Retinal structural and functional changes in humans can be manifestations of different physiological or pathological conditions. Retinal imaging is the only way to directly inspect blood vessels and their pathological changes throughout the whole body non-invasively. Various quantitative analysis metrics have been used to measure the abnormalities of retinal microvasculature in the context of different retinal, cerebral and systemic disorders. Recently developed optical coherence tomography angiography (OCTA) is a non-invasive imaging tool that allows high-resolution three-dimensional mapping of the retinal microvasculature. The identification of retinal biomarkers from OCTA images could facilitate clinical investigation in various scenarios. We provide a framework for extracting computational retinal microvasculature biomarkers (CRMBs) from OCTA images through a knowledge-driven computerized automatic analytical system. Our method allows for improved identification of the foveal avascular zone (FAZ) and introduces a novel definition of vessel dispersion in the macular region. Furthermore, retinal large vessels and capillaries of the superficial and deep plexus can be differentiated, correlating with retinal pathology. The diagnostic value of OCTA CRMBs was demonstrated by a cross-sectional study with 30 healthy subjects and 43 retinal vein occlusion (RVO) patients, which identified strong correlations between OCTA CRMBs and retinal function in RVO patients. These OCTA CRMBs generated through this "all-in-one" pipeline may provide clinicians with insights about disease severity, treatment response and prognosis, aiding in the management and early detection of various disorders.
Collapse
Affiliation(s)
- Bingwen Lu
- Department of Ophthalmology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China;
- Department of Ophthalmology, Eye Hospital, China Academy of Chinese Medical Sciences, Beijing 100040, China
| | - Yiming Li
- Department of Psychiatry, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (Y.L.); (P.-C.S.)
- Centre for PanorOmic Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Like Xie
- Department of Ophthalmology, Eye Hospital, China Academy of Chinese Medical Sciences, Beijing 100040, China
| | - Kin Chiu
- Department of Ophthalmology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China;
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong SAR, China
- Department of Psychology, The University of Hong Kong, Hong Kong SAR, China
| | - Xiaofeng Hao
- Department of Ophthalmology, Eye Hospital, China Academy of Chinese Medical Sciences, Beijing 100040, China
| | - Jing Xu
- Department of Ophthalmology, Eye Hospital, China Academy of Chinese Medical Sciences, Beijing 100040, China
| | - Jie Luo
- Department of Ophthalmology, Eye Hospital, China Academy of Chinese Medical Sciences, Beijing 100040, China
| | - Pak-Chung Sham
- Department of Psychiatry, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (Y.L.); (P.-C.S.)
- Centre for PanorOmic Sciences, The University of Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
12
|
Yu H, Wang X, Tian R, Li X, Xu C, Fei J, Li T, Yin Z. Myometrium infection decreases TREK1 through NHE1 and increases contraction in pregnant mice. Am J Physiol Cell Physiol 2024; 326:C1106-C1119. [PMID: 38344766 DOI: 10.1152/ajpcell.00598.2023] [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: 11/08/2023] [Revised: 02/07/2024] [Accepted: 02/08/2024] [Indexed: 03/23/2024]
Abstract
Intrauterine infection during pregnancy can enhance uterine contractions. A two-pore K+ channel TREK1 is crucial for maintaining uterine quiescence and reducing contractility, with its properties regulated by pH changes in cell microenvironment. Meanwhile, the sodium hydrogen exchanger 1 (NHE1) plays a pivotal role in modulating cellular pH homeostasis, and its activation increases smooth muscle tension. By establishing an infected mouse model of Escherichia coli (E. coli) and lipopolysaccharide (LPS), we used Western blotting, real-time quantitative polymerase chain reaction, and immunofluorescence to detect changes of TREK1 and NHE1 expression in the myometrium, and isometric recording measured the uterus contraction. The NHE1 inhibitor cariporide was used to explore the effect of NHE1 on TREK1. Finally, cell contraction assay and siRNA transfection were performed to clarify the relationship between NHE1 and TREK1 in vitro. We found that the uterine contraction was notably enhanced in infected mice with E. coli and LPS administration. Meanwhile, TREK1 expression was reduced, whereas NHE1 expression was upregulated in infected mice. Cariporide alleviated the increased uterine contraction and promoted myometrium TREK1 expression in LPS-injected mice. Furthermore, suppression of NHE1 with siRNA transfection inhibited the contractility of uterine smooth muscle cells and activated the TREK1. Altogether, our findings indicate that infection increases the uterine contraction by downregulating myometrium TREK1 in mice, and the inhibition of TREK1 is attributed to the activation of NHE1.NEW & NOTEWORTHY Present work found that infection during pregnancy will increase myometrium contraction. Infection downregulated NHE1 and followed TREK1 expression and activation decrease in myometrium, resulting in increased myometrium contraction.
Collapse
Affiliation(s)
- Huihui Yu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xingxing Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ruixian Tian
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xuan Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chenyi Xu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jiajia Fei
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Tengteng Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zongzhi Yin
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, China
| |
Collapse
|
13
|
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
|
14
|
McDonald H, Gardner-Russell J, Alarcon-Martinez L. Orchestrating Blood Flow in the Retina: Interpericyte Tunnelling Nanotube Communication. Results Probl Cell Differ 2024; 73:229-247. [PMID: 39242382 DOI: 10.1007/978-3-031-62036-2_11] [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] [Indexed: 09/09/2024]
Abstract
The retina transforms light into electrical signals, which are sent to the brain via the optic nerve to form our visual perception. This complex signal processing is performed by the retinal neuron and requires a significant amount of energy. Since neurons are unable to store energy, they must obtain glucose and oxygen from the bloodstream to produce energy to match metabolic needs. This process is called neurovascular coupling (NVC), and it is based on a precise mechanism that is not totally understood. The discovery of fine tubular processes termed tunnelling nanotubes (TNTs) set a new type of cell-to-cell communication. TNTs are extensions of the cellular membrane that allow the transfer of material between connected cells. Recently, they have been reported in the brain and retina of living mice, where they connect pericytes, which are vascular mural cells that regulate vessel diameter. Accordingly, these TNTs were termed interpericyte tunnelling nanotubes (IPTNTs), which showed a vital role in blood delivery and NVC. In this chapter, we review the involvement of TNTs in NVC and discuss their implications in retinal neurodegeneration.
Collapse
Affiliation(s)
- Hannah McDonald
- Centre for Eye Research Australia, Melbourne, VIC, Australia
- Department of Ophthalmology, University of Melbourne, Melbourne, VIC, Australia
- The Royal Victorian Eye and Ear Hospital, Melbourne, VIC, Australia
| | - Jesse Gardner-Russell
- Centre for Eye Research Australia, Melbourne, VIC, Australia
- Department of Ophthalmology, University of Melbourne, Melbourne, VIC, Australia
- The Royal Victorian Eye and Ear Hospital, Melbourne, VIC, Australia
| | - Luis Alarcon-Martinez
- Centre for Eye Research Australia, Melbourne, VIC, Australia.
- Department of Ophthalmology, University of Melbourne, Melbourne, VIC, Australia.
- The Royal Victorian Eye and Ear Hospital, Melbourne, VIC, Australia.
| |
Collapse
|
15
|
Iba C, Abe Y, Tanaka KF. Optogenetic demonstration of the involvement of SMA-negative mural cells in the regulation of cerebral blood flow. Front Physiol 2023; 14:1322250. [PMID: 38187133 PMCID: PMC10771846 DOI: 10.3389/fphys.2023.1322250] [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: 10/16/2023] [Accepted: 12/08/2023] [Indexed: 01/09/2024] Open
Abstract
Mural cells are critical components of the cerebral vasculature. They are categorized into three primary subsets: arteriole smooth muscle cells (aSMCs), pericytes (PCs) and venule smooth muscle cells (vSMCs). It is well known that aSMCs can directly regulate cerebral blood flow (CBF) with their own contraction and dilation mechanisms. On the other hand, the direct involvement of PCs or vSMCs in CBF regulation is controversial. This ambiguity is largely due to the lack of specifically manipulable tools to isolate their function. To address this issue, we employed a set-subtraction approach by using a combination of tTA-mediated gene induction and Cre-mediated gene excision. We developed transgenic mice expressing optical actuators, channelrhodopsin-2 (ChR2) and photoactivated adenylyl cyclase (PAC) in smooth muscle actin (SMA)-negative mural cells that lack the machinery for SMA-mediated vasoregulation. Using these mouse models, we assessed CBF alterations in response to optical stimulation using laser Doppler techniques. Our results showed that optical stimulation induced notable CBF changes in both models. This study provides evidence for the potential regulatory role of PCs and vSMCs in cerebral hemodynamics and introduces powerful tools to specifically manipulate these cell types in vascular neurobiology.
Collapse
Affiliation(s)
| | | | - Kenji F. Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
16
|
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: 26] [Impact Index Per Article: 13.0] [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
|
17
|
Xu S, Li S, Yan Z, Wang Y, Zhang L. Development and Validation of a UHPLC-MS/MS Method for the Quantification of a Novel PYGB Inhibitor in Plasma: Application to Pharmacokinetic Studies. Molecules 2023; 28:6995. [PMID: 37836837 PMCID: PMC10574475 DOI: 10.3390/molecules28196995] [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: 08/11/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
In previous studies, we reported compound 1 (5-chloro-N-(4-oxo-2,2-dipropyl-3,4-dihydro-2H-benzo[e][1,3]oxazin-6-yl)-1H-indole-2-carboxamide) as a novel PYGB inhibitor, and found that it had better anti-ischemic brain injury activity. In this study, we established and validated a novel UHPLC-MS/MS method for the quantitative determination of compound 1 in plasma, then applied the method to study the pharmacokinetic parameters and brain tissue distribution of compound 1 in SD (Sprague-Dawley) rats after intravenous administration. The experimental results showed that the method met the validation requirements set by the US FDA in terms of linearity, accuracy, precision, and stability. The validated method was then used for pharmacokinetic studies in rat plasma, and it was found that compound 1 exhibited linear pharmacokinetic characteristics when administered in the dose range of 0.8-3.2 mg/kg. Finally, we also conducted a brief preliminary investigation of the brain tissue distribution of compound 1 in rats after injection and found that the brain tissue concentrations at 0.25 h and 2 h of administration were 440 ± 19.1 ng/kg and 111 ± 23.9 ng/kg, respectively. Additionally, the CBrain/CPlasma ratio was 0.112 ± 0.0185 and 0.112 ± 0.0292, respectively. These results indicated that compound 1 was able to cross the blood-brain barrier. This study provides important support for the application of compound 1 in ischemic brain injury diseases.
Collapse
Affiliation(s)
| | | | | | | | - Liying Zhang
- Laboratory of Traditional Chinese Medicine Research and Development of Hebei Province, Institute of Traditional Chinese Medicine, Chengde Medical University, Chengde 067000, China; (S.X.); (S.L.); (Z.Y.)
| |
Collapse
|
18
|
Wei J, Zhang L, Wu K, Yu J, Gao F, Cheng J, Zhang T, Zhou X, Zong Y, Huang X, Jiang C. R-(+)-WIN55212-2 protects pericytes from ischemic damage and restores retinal microcirculatory patency after ischemia/reperfusion injury. Biomed Pharmacother 2023; 166:115197. [PMID: 37572634 DOI: 10.1016/j.biopha.2023.115197] [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: 04/14/2023] [Revised: 07/09/2023] [Accepted: 07/18/2023] [Indexed: 08/14/2023] Open
Abstract
BACKGROUND AND PURPOSE Cannabinoids are vasoactive substances that act as key regulators of arterial tone in the blood vessels supplying peripheral tissues and the central nervous system. This study aimed to investigate the potential of R-(+)-WIN55212-2 (WIN), a cannabinoid receptor 1 agonist (CB1), as a treatment for retinal ischemia/reperfusion (I/R) injury. EXPERIMENTAL APPROACH Male Wistar rats were subjected to retinal I/R injury by increasing intraocular pressure in the anterior chamber. The rats were randomly divided into four groups: normal control, I/R, vehicle (pre-treated with dimethyl sulfoxide [DMSO] via intraperitoneal injection), and experimental (pre-treated with WIN at a dose of 1 ml/kg via intraperitoneal injection). The rats were sacrificed at different time points of reperfusion (1 hour, 3 hours, 6 hours, and 1 day) after inducing retinal I/R injury, and their retinas were collected for analysis. Oxygen-glucose deprived/reperfusion (OGD/R) was performed by initially perfusing the retinas with oxygenated artificial cerebrospinal fluid (ACSF), then switching to an OGD solution to simulate ischemia, followed by another perfusion with ACSF. Pericyte contraction and the "no-reflow" phenomenon were observed using infrared differential interference contrast (IR-DIC) microscopy and immunohistochemistry. Western blot, enzyme-linked immunosorbent assay (ELISA), and nitric oxide (NO) detection were used to explore the potential mechanism. KEY RESULTS In both the OGD/R and I/R models, retinal pericytes exhibited persistent contraction even after reperfusion. The ability of WIN to regulate the tone of retinal pericytes and capillaries was specifically blocked by the BKCa inhibitor iberiotoxin (100 nM). WIN demonstrated a protective effect against retinal I/R injury by preserving blood flow in vessels containing pericytes. Pretreatment with WIN alleviated the persistent contraction and apoptosis of retinal pericytes in I/R-induced rats, accompanied by a reduction in intracellular calcium ion (Ca2+) concentration. The expression of CB1 decreased in a time-dependent manner in the I/R group. After I/R injury, endothelium-derived nitric oxide (eNOS) levels were reduced at all time points, which was successfully reversed by WIN therapy except for the 1 day group. Additionally, the downregulation of cyclic guanosine monophosphate (cGMP) and BKCa expression at 3 hours, 6 hours, and 1 day after I/R injury was restored by pretreatment of WIN. CONCLUSIONS & IMPLICATIONS WIN exerted its protective effects on retinal I/R injury by inhibiting the contraction and apoptosis of pericytes through the CB1-eNOS-cGMP-BKCa signaling pathway, thus ameliorated the occlusion of retinal capillaries.
Collapse
Affiliation(s)
- Jiaojiao Wei
- Department of Ophthalmology and Vision Science, Eye and ENT Hospital, Fudan University, Shanghai 200031, People's Republic of China; Eye and ENT Hospital, State Key laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200031, People's Republic of China; Key Laboratory of Myopia of State Health Ministry, and Key Laboratory of Visual Impairment and Restoration of Shanghai, Shanghai 200031, People's Republic of China
| | - Lili Zhang
- Department of Ophthalmology and Vision Science, Eye and ENT Hospital, Fudan University, Shanghai 200031, People's Republic of China; Eye and ENT Hospital, State Key laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200031, People's Republic of China; Key Laboratory of Myopia of State Health Ministry, and Key Laboratory of Visual Impairment and Restoration of Shanghai, Shanghai 200031, People's Republic of China
| | - Kaicheng Wu
- Department of Ophthalmology and Vision Science, Eye and ENT Hospital, Fudan University, Shanghai 200031, People's Republic of China; Eye and ENT Hospital, State Key laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200031, People's Republic of China; Key Laboratory of Myopia of State Health Ministry, and Key Laboratory of Visual Impairment and Restoration of Shanghai, Shanghai 200031, People's Republic of China
| | - Jian Yu
- Department of Ophthalmology and Vision Science, Eye and ENT Hospital, Fudan University, Shanghai 200031, People's Republic of China; Eye and ENT Hospital, State Key laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200031, People's Republic of China; Key Laboratory of Myopia of State Health Ministry, and Key Laboratory of Visual Impairment and Restoration of Shanghai, Shanghai 200031, People's Republic of China
| | - Fengjuan Gao
- Department of Ophthalmology and Vision Science, Eye and ENT Hospital, Fudan University, Shanghai 200031, People's Republic of China; Eye and ENT Hospital, State Key laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200031, People's Republic of China; Key Laboratory of Myopia of State Health Ministry, and Key Laboratory of Visual Impairment and Restoration of Shanghai, Shanghai 200031, People's Republic of China
| | - Jingyi Cheng
- Department of Ophthalmology and Vision Science, Eye and ENT Hospital, Fudan University, Shanghai 200031, People's Republic of China; Eye and ENT Hospital, State Key laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200031, People's Republic of China; Key Laboratory of Myopia of State Health Ministry, and Key Laboratory of Visual Impairment and Restoration of Shanghai, Shanghai 200031, People's Republic of China
| | - Ting Zhang
- Department of Ophthalmology and Vision Science, Eye and ENT Hospital, Fudan University, Shanghai 200031, People's Republic of China; Eye and ENT Hospital, State Key laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200031, People's Republic of China; Key Laboratory of Myopia of State Health Ministry, and Key Laboratory of Visual Impairment and Restoration of Shanghai, Shanghai 200031, People's Republic of China
| | - Xujiao Zhou
- Eye and ENT Hospital, State Key laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200031, People's Republic of China; Key Laboratory of Myopia of State Health Ministry, and Key Laboratory of Visual Impairment and Restoration of Shanghai, Shanghai 200031, People's Republic of China.
| | - Yuan Zong
- Department of Ophthalmology and Vision Science, Eye and ENT Hospital, Fudan University, Shanghai 200031, People's Republic of China; Eye and ENT Hospital, State Key laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200031, People's Republic of China; Key Laboratory of Myopia of State Health Ministry, and Key Laboratory of Visual Impairment and Restoration of Shanghai, Shanghai 200031, People's Republic of China.
| | - Xiaojing Huang
- Department of Ophthalmology, Shanghai Pudong New Area Gongli Hospital, Shanghai 200135, People's Republic of China.
| | - Chunhui Jiang
- Department of Ophthalmology and Vision Science, Eye and ENT Hospital, Fudan University, Shanghai 200031, People's Republic of China; Eye and ENT Hospital, State Key laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200031, People's Republic of China; Key Laboratory of Myopia of State Health Ministry, and Key Laboratory of Visual Impairment and Restoration of Shanghai, Shanghai 200031, People's Republic of China
| |
Collapse
|
19
|
Fu J, Liang H, Yuan P, Wei Z, Zhong P. Brain pericyte biology: from physiopathological mechanisms to potential therapeutic applications in ischemic stroke. Front Cell Neurosci 2023; 17:1267785. [PMID: 37780206 PMCID: PMC10536258 DOI: 10.3389/fncel.2023.1267785] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 08/30/2023] [Indexed: 10/03/2023] Open
Abstract
Pericytes play an indispensable role in various organs and biological processes, such as promoting angiogenesis, regulating microvascular blood flow, and participating in immune responses. Therefore, in this review, we will first introduce the discovery and development of pericytes, identification methods and functional characteristics, then focus on brain pericytes, on the one hand, to summarize the functions of brain pericytes under physiological conditions, mainly discussing from the aspects of stem cell characteristics, contractile characteristics and paracrine characteristics; on the other hand, to summarize the role of brain pericytes under pathological conditions, mainly taking ischemic stroke as an example. Finally, we will discuss and analyze the application and development of pericytes as therapeutic targets, providing the research basis and direction for future microvascular diseases, especially ischemic stroke treatment.
Collapse
Affiliation(s)
- Jiaqi Fu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- Department of Neurology, Shidong Hospital, Yangpu District, Shanghai, China
| | - Huazheng Liang
- Monash Suzhou Research Institute, Suzhou, Jiangsu, China
| | - Ping Yuan
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhenyu Wei
- Department of Neurology, Shidong Hospital, Yangpu District, Shanghai, China
| | - Ping Zhong
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- Department of Neurology, Shidong Hospital, Yangpu District, Shanghai, China
| |
Collapse
|
20
|
Gurler G, Belder N, Beker MC, Sever-Bahcekapili M, Uruk G, Kilic E, Yemisci M. Reduced folate carrier 1 is present in retinal microvessels and crucial for the inner blood retinal barrier integrity. Fluids Barriers CNS 2023; 20:47. [PMID: 37328777 DOI: 10.1186/s12987-023-00442-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 05/18/2023] [Indexed: 06/18/2023] Open
Abstract
BACKGROUND Reduced folate carrier 1 (RFC1; SLC19a1) is the main responsible transporter for the B9 family of vitamins named folates, which are essential for normal tissue growth and development. While folate deficiency resulted in retinal vasculopathy, the expression and the role of RFC1 in blood-retinal barrier (BRB) are not well known. METHODS We used whole mount retinas and trypsin digested microvessel samples of adult mice. To knockdown RFC1, we delivered RFC1-targeted short interfering RNA (RFC1-siRNA) intravitreally; while, to upregulate RFC1 we delivered lentiviral vector overexpressing RFC1. Retinal ischemia was induced 1-h by applying FeCl3 to central retinal artery. We used RT-qPCR and Western blotting to determine RFC1. Endothelium (CD31), pericytes (PDGFR-beta, CD13, NG2), tight-junctions (Occludin, Claudin-5 and ZO-1), main basal membrane protein (Collagen-4), endogenous IgG and RFC1 were determined immunohistochemically. RESULTS Our analyses on whole mount retinas and trypsin digested microvessel samples of adult mice revealed the presence of RFC1 in the inner BRB and colocalization with endothelial cells and pericytes. Knocking down RFC1 expression via siRNA delivery resulted in the disintegration of tight junction proteins and collagen-4 in twenty-four hours, which was accompanied by significant endogenous IgG extravasation. This indicated the impairment of BRB integrity after an abrupt RFC1 decrease. Furthermore, lentiviral vector-mediated RFC1 overexpression resulted in increased tight junction proteins and collagen-4, confirming the structural role of RFC1 in the inner BRB. Acute retinal ischemia decreased collagen-4 and occludin levels and led to an increase in RFC1. Besides, the pre-ischemic overexpression of RFC1 partially rescued collagen-4 and occludin levels which would be decreased after ischemia. CONCLUSION In conclusion, our study clarifies the presence of RFC1 protein in the inner BRB, which has recently been defined as hypoxia-immune-related gene in other tissues and offers a novel perspective of retinal RFC1. Hence, other than being a folate carrier, RFC1 is an acute regulator of the inner BRB in healthy and ischemic retinas.
Collapse
Affiliation(s)
- Gokce Gurler
- The Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| | - Nevin Belder
- Biotechnology Institute, Ankara University, Ankara, Turkey
| | | | | | - Gokhan Uruk
- The Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| | - Ertugrul Kilic
- Neuroscience and Neurotechnology Center of Excellence (NÖROM), Ankara, Turkey
- Physiology, Istanbul Medeniyet University, Istanbul, Turkey
| | - Muge Yemisci
- The Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey.
- Faculty of Medicine, Department of Neurology, Hacettepe University, Ankara, Turkey.
- Neuroscience and Neurotechnology Center of Excellence (NÖROM), Ankara, Turkey.
| |
Collapse
|
21
|
Mechanisms of the "No-Reflow" Phenomenon After Acute Myocardial Infarction: Potential Role of Pericytes. JACC Basic Transl Sci 2023; 8:204-220. [PMID: 36908667 PMCID: PMC9998747 DOI: 10.1016/j.jacbts.2022.06.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/13/2022] [Accepted: 06/13/2022] [Indexed: 11/20/2022]
Abstract
Pericytes contract during myocardial ischemia resulting in capillary constriction and no reflow. Reversing pericyte contraction pharmacologically reduces no reflow and infarct size. These findings open up an entire new venue of research aimed at altering pericyte function in myocardial ischemia and infarction.
Collapse
|
22
|
Haider AA, Rex TS, Wareham LK. cGMP Signaling in the Neurovascular Unit-Implications for Retinal Ganglion Cell Survival in Glaucoma. Biomolecules 2022; 12:1671. [PMID: 36421684 PMCID: PMC9687235 DOI: 10.3390/biom12111671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Glaucoma is a progressive age-related disease of the visual system and the leading cause of irreversible blindness worldwide. Currently, intraocular pressure (IOP) is the only modifiable risk factor for the disease, but even as IOP is lowered, the pathology of the disease often progresses. Hence, effective clinical targets for the treatment of glaucoma remain elusive. Glaucoma shares comorbidities with a multitude of vascular diseases, and evidence in humans and animal models demonstrates an association between vascular dysfunction of the retina and glaucoma pathology. Integral to the survival of retinal ganglion cells (RGCs) is functional neurovascular coupling (NVC), providing RGCs with metabolic support in response to neuronal activity. NVC is mediated by cells of the neurovascular unit (NVU), which include vascular cells, glial cells, and neurons. Nitric oxide-cyclic guanosine monophosphate (NO-cGMP) signaling is a prime mediator of NVC between endothelial cells and neurons, but emerging evidence suggests that cGMP signaling is also important in the physiology of other cells of the NVU. NO-cGMP signaling has been implicated in glaucomatous neurodegeneration in humans and mice. In this review, we explore the role of cGMP signaling in the different cell types of the NVU and investigate the potential links between cGMP signaling, breakdown of neurovascular function, and glaucoma pathology.
Collapse
Affiliation(s)
| | | | - Lauren K. Wareham
- Vanderbilt Eye Institute, Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, TN 37212, USA
| |
Collapse
|
23
|
Tomanek RJ. The coronary capillary bed and its role in blood flow and oxygen delivery: A review. Anat Rec (Hoboken) 2022; 305:3199-3211. [PMID: 35521832 PMCID: PMC9796134 DOI: 10.1002/ar.24951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/19/2022] [Accepted: 04/21/2022] [Indexed: 01/01/2023]
Abstract
The assumption that the coronary capillary blood flow is exclusively regulated by precapillary vessels is not supported by recent data. Rather, the complex coronary capillary bed has unique structural and geometric characteristics that invalidate many assumptions regarding red blood cell (RBC) transport, for example, data based on a single capillary or that increases in flow are the result of capillary recruitment. It is now recognized that all coronary capillaries are open and that their variations in flow are due to structural differences, local O2 demand and delivery, and variations in hematocrit. Recent data reveal that local mechanisms within the capillary bed regulate flow via signaling mechanisms involving RBC signaling and endothelial-associated pericytes that contract and relax in response to humoral and neural signaling. The discovery that pericytes respond to vasoactive signals (e.g., nitric oxide, phenylephrine, and adenosine) underscores the role of these cells in regulating capillary diameter and consequently RBC flux and oxygen delivery. RBCs also affect blood flow by sensing <mml:math xmlns:mml="http://www.w3.org/1998/Math/MathML"><mml:msub><mml:mi>P</mml:mi> <mml:msub><mml:mi>O</mml:mi> <mml:mn>2</mml:mn></mml:msub> </mml:msub> </mml:math> and releasing nitric oxide to facilitate relaxation of pericytes and a consequential capillary dilation. New data indicate that these signaling mechanisms allow control of blood flow in specific coronary capillaries according to their oxygen requirements. In conclusion, mechanisms in the coronary capillary bed facilitate RBC density and transit time, hematocrit, blood flow and O2 delivery, factors that decrease capillary heterogeneity. These findings have important clinical implications for myocardial ischemia and infarction, as well as other vascular diseases.
Collapse
Affiliation(s)
- Robert J. Tomanek
- Department of Anatomy and Cell Biology, Carver College of MedicineUniversity of IowaIowa CityIAUSA
| |
Collapse
|
24
|
Balaratnasingam C, An D, Hein M, Yu P, Yu DY. Studies of the retinal microcirculation using human donor eyes and high-resolution clinical imaging: Insights gained to guide future research in diabetic retinopathy. Prog Retin Eye Res 2022; 94:101134. [PMID: 37154065 DOI: 10.1016/j.preteyeres.2022.101134] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 09/18/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022]
Abstract
The microcirculation plays a key role in delivering oxygen to and removing metabolic wastes from energy-intensive retinal neurons. Microvascular changes are a hallmark feature of diabetic retinopathy (DR), a major cause of irreversible vision loss globally. Early investigators have performed landmark studies characterising the pathologic manifestations of DR. Previous works have collectively informed us of the clinical stages of DR and the retinal manifestations associated with devastating vision loss. Since these reports, major advancements in histologic techniques coupled with three-dimensional image processing has facilitated a deeper understanding of the structural characteristics in the healthy and diseased retinal circulation. Furthermore, breakthroughs in high-resolution retinal imaging have facilitated clinical translation of histologic knowledge to detect and monitor progression of microcirculatory disturbances with greater precision. Isolated perfusion techniques have been applied to human donor eyes to further our understanding of the cytoarchitectural characteristics of the normal human retinal circulation as well as provide novel insights into the pathophysiology of DR. Histology has been used to validate emerging in vivo retinal imaging techniques such as optical coherence tomography angiography. This report provides an overview of our research on the human retinal microcirculation in the context of the current ophthalmic literature. We commence by proposing a standardised histologic lexicon for characterising the human retinal microcirculation and subsequently discuss the pathophysiologic mechanisms underlying key manifestations of DR, with a focus on microaneurysms and retinal ischaemia. The advantages and limitations of current retinal imaging modalities as determined using histologic validation are also presented. We conclude with an overview of the implications of our research and provide a perspective on future directions in DR research.
Collapse
Affiliation(s)
- Chandrakumar Balaratnasingam
- Lions Eye Institute, Nedlands, Western Australia, Australia; Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Australia; Department of Ophthalmology, Sir Charles Gairdner Hospital, Western Australia, Australia.
| | - Dong An
- Lions Eye Institute, Nedlands, Western Australia, Australia; Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Australia
| | - Martin Hein
- Lions Eye Institute, Nedlands, Western Australia, Australia; Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Australia
| | - Paula Yu
- Lions Eye Institute, Nedlands, Western Australia, Australia; Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Australia
| | - Dao-Yi Yu
- Lions Eye Institute, Nedlands, Western Australia, Australia; Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Australia
| |
Collapse
|
25
|
Guo RB, Dong YF, Yin Z, Cai ZY, Yang J, Ji J, Sun YQ, Huang XX, Xue TF, Cheng H, Zhou XQ, Sun XL. Iptakalim improves cerebral microcirculation in mice after ischemic stroke by inhibiting pericyte contraction. Acta Pharmacol Sin 2022; 43:1349-1359. [PMID: 34697419 PMCID: PMC9160281 DOI: 10.1038/s41401-021-00784-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 09/23/2021] [Indexed: 02/07/2023]
Abstract
Pericytes are present tight around the intervals of capillaries, play an essential role in stabilizing the blood-brain barrier, regulating blood flow and immunomodulation, and persistent contraction of pericytes eventually leads to impaired blood flow and poor clinical outcomes in ischemic stroke. We previously show that iptakalim, an ATP-sensitive potassium (K-ATP) channel opener, exerts protective effects in neurons, and glia against ischemia-induced injury. In this study we investigated the impacts of iptakalim on pericytes contraction in stroke. Mice were subjected to cerebral artery occlusion (MCAO), then administered iptakalim (10 mg/kg, ip). We showed that iptakalim administration significantly promoted recovery of cerebral blood flow after cerebral ischemia and reperfusion. Furthermore, we found that iptakalim significantly inhibited pericytes contraction, decreased the number of obstructed capillaries, and improved cerebral microcirculation. Using a collagen gel contraction assay, we demonstrated that cultured pericytes subjected to oxygen-glucose deprivation (OGD) consistently contracted from 3 h till 24 h during reoxygenation, whereas iptakalim treatment (10 μM) notably restrained pericyte contraction from 6 h during reoxygenation. We further showed that iptakalim treatment promoted K-ATP channel opening via suppressing SUR2/EPAC1 complex formation. Consequently, it reduced calcium influx and ET-1 release. Taken together, our results demonstrate that iptakalim, targeted K-ATP channels, can improve microvascular disturbance by inhibiting pericyte contraction after ischemic stroke. Our work reveals that iptakalim might be developed as a promising pericyte regulator for treatment of stroke.
Collapse
Affiliation(s)
- Ruo-bing Guo
- grid.89957.3a0000 0000 9255 8984Neuroprotective Drug Discovery Key Laboratory, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, 211166 China
| | - Yin-feng Dong
- grid.410745.30000 0004 1765 1045Nanjing University of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029 China
| | - Zhi Yin
- grid.412676.00000 0004 1799 0784The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| | - Zhen-yu Cai
- grid.89957.3a0000 0000 9255 8984Neuroprotective Drug Discovery Key Laboratory, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, 211166 China
| | - Jin Yang
- grid.89957.3a0000 0000 9255 8984Neuroprotective Drug Discovery Key Laboratory, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, 211166 China
| | - Juan Ji
- grid.89957.3a0000 0000 9255 8984Neuroprotective Drug Discovery Key Laboratory, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, 211166 China
| | - Yu-qin Sun
- grid.89957.3a0000 0000 9255 8984Neuroprotective Drug Discovery Key Laboratory, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, 211166 China
| | - Xin-xin Huang
- grid.412676.00000 0004 1799 0784The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| | - Teng-fei Xue
- grid.89957.3a0000 0000 9255 8984Neuroprotective Drug Discovery Key Laboratory, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, 211166 China
| | - Hong Cheng
- grid.412676.00000 0004 1799 0784The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| | - Xi-qiao Zhou
- grid.410745.30000 0004 1765 1045Nanjing University of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029 China
| | - Xiu-lan Sun
- grid.89957.3a0000 0000 9255 8984Neuroprotective Drug Discovery Key Laboratory, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, 211166 China ,grid.410745.30000 0004 1765 1045Nanjing University of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029 China
| |
Collapse
|
26
|
Girolamo F, Errede M, Bizzoca A, Virgintino D, Ribatti D. Central Nervous System Pericytes Contribute to Health and Disease. Cells 2022; 11:1707. [PMID: 35626743 PMCID: PMC9139243 DOI: 10.3390/cells11101707] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/16/2022] [Accepted: 05/18/2022] [Indexed: 12/11/2022] Open
Abstract
Successful neuroprotection is only possible with contemporary microvascular protection. The prevention of disease-induced vascular modifications that accelerate brain damage remains largely elusive. An improved understanding of pericyte (PC) signalling could provide important insight into the function of the neurovascular unit (NVU), and into the injury-provoked responses that modify cell-cell interactions and crosstalk. Due to sharing the same basement membrane with endothelial cells, PCs have a crucial role in the control of endothelial, astrocyte, and oligodendrocyte precursor functions and hence blood-brain barrier stability. Both cerebrovascular and neurodegenerative diseases impair oxygen delivery and functionally impair the NVU. In this review, the role of PCs in central nervous system health and disease is discussed, considering their origin, multipotency, functions and also dysfunction, focusing on new possible avenues to modulate neuroprotection. Dysfunctional PC signalling could also be considered as a potential biomarker of NVU pathology, allowing us to individualize therapeutic interventions, monitor responses, or predict outcomes.
Collapse
Affiliation(s)
- Francesco Girolamo
- Unit of Human Anatomy and Histology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro’, 70124 Bari, Italy; (M.E.); (D.V.); (D.R.)
| | - Mariella Errede
- Unit of Human Anatomy and Histology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro’, 70124 Bari, Italy; (M.E.); (D.V.); (D.R.)
| | - Antonella Bizzoca
- Physiology Unit, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro’, 70124 Bari, Italy;
| | - Daniela Virgintino
- Unit of Human Anatomy and Histology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro’, 70124 Bari, Italy; (M.E.); (D.V.); (D.R.)
| | - Domenico Ribatti
- Unit of Human Anatomy and Histology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro’, 70124 Bari, Italy; (M.E.); (D.V.); (D.R.)
| |
Collapse
|
27
|
Schreiner TG, Romanescu C, Popescu BO. The Blood-Brain Barrier-A Key Player in Multiple Sclerosis Disease Mechanisms. Biomolecules 2022; 12:538. [PMID: 35454127 PMCID: PMC9025898 DOI: 10.3390/biom12040538] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/28/2022] [Accepted: 03/30/2022] [Indexed: 02/07/2023] Open
Abstract
Over the past decade, multiple sclerosis (MS), a chronic neuroinflammatory disease with severe personal and social consequences, has undergone a steady increase in incidence and prevalence rates worldwide. Despite ongoing research and the development of several novel therapies, MS pathology remains incompletely understood, and the prospect for a curative treatment continues to be unpromising in the near future. A sustained research effort, however, should contribute to a deeper understanding of underlying disease mechanisms, which will undoubtedly yield improved results in drug development. In recent years, the blood-brain barrier (BBB) has increasingly become the focus of many studies as it appears to be involved in both MS disease onset and progression. More specifically, neurovascular unit damage is believed to be involved in the critical process of CNS immune cell penetration, which subsequently favors the development of a CNS-specific immune response, leading to the classical pathological and clinical hallmarks of MS. The aim of the current narrative review is to merge the relevant evidence on the role of the BBB in MS pathology in a comprehensive and succinct manner. Firstly, the physiological structure and functions of the BBB as a component of the more complex neurovascular unit are presented. Subsequently, the authors review the specific alteration of the BBB encountered in different stages of MS, focusing on both the modifications of BBB cells in neuroinflammation and the CNS penetration of immune cells. Finally, the currently accepted theories on neurodegeneration in MS are summarized.
Collapse
Affiliation(s)
- Thomas Gabriel Schreiner
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
- Faculty of Medicine, “Gr. T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
- Department of Electrical Measurements and Materials, Faculty of Electrical Engineering and Information Technology, Gheorghe Asachi Technical University of Iasi, 21-23 Professor Dimitrie Mangeron Blvd., 700050 Iasi, Romania
| | - Constantin Romanescu
- Faculty of Medicine, “Gr. T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
- Clinical Section IV, “St. Parascheva” Infectious Disease Hospital, 700116 Iași, Romania
| | - Bogdan Ovidiu Popescu
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
- Neurology Department, Colentina Clinical Hospital, 020125 Bucharest, Romania
- Laboratory of Cell Biology, Neurosciences and Experimental Myology, “Victor Babes” National Institute of Pathology, 050096 Bucharest, Romania
| |
Collapse
|
28
|
Erdener ŞE, Küreli G, Dalkara T. Contractile apparatus in CNS capillary pericytes. NEUROPHOTONICS 2022; 9:021904. [PMID: 35106320 PMCID: PMC8785978 DOI: 10.1117/1.nph.9.2.021904] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 12/22/2021] [Indexed: 06/14/2023]
Abstract
Significance: Whether or not capillary pericytes contribute to blood flow regulation in the brain and retina has long been debated. This was partly caused by failure of detecting the contractile protein α -smooth muscle actin ( α -SMA) in capillary pericytes. Aim: The aim of this review is to summarize recent developments in detecting α -SMA and contractility in capillary pericytes and the relevant literature on the biology of actin filaments. Results: Evidence suggests that for visualization of the small amounts of α -SMA in downstream mid-capillary pericytes, actin depolymerization must be prevented during tissue processing. Actin filaments turnover is mainly based on de/re-polymerization rather than transcription of the monomeric form, hence, small amounts of α -SMA mRNA may evade detection by transcriptomic studies. Similarly, transgenic mice expressing fluorescent reporters under the α -SMA promoter may yield low fluorescence due to limited transcriptional activity in mid-capillary pericytes. Recent studies show that pericytes including mid-capillary ones express several actin isoforms and myosin heavy chain type 11, the partner of α -SMA in mediating contraction. Emerging evidence also suggests that actin polymerization in pericytes may have a role in regulating the tone of downstream capillaries. Conclusions: With guidance of actin biology, innovative labeling and imaging techniques can reveal the molecular machinery of contraction in pericytes.
Collapse
Affiliation(s)
- Şefik E. Erdener
- Hacettepe University, Institute of Neurological Sciences and Psychiatry, Ankara, Turkey
| | - Gülce Küreli
- Hacettepe University, Institute of Neurological Sciences and Psychiatry, Ankara, Turkey
| | - Turgay Dalkara
- Hacettepe University, Institute of Neurological Sciences and Psychiatry, Ankara, Turkey
| |
Collapse
|
29
|
Freitas F, Attwell D. Pericyte-mediated constriction of renal capillaries evokes no-reflow and kidney injury following ischaemia. eLife 2022; 11:74211. [PMID: 35285797 PMCID: PMC8947765 DOI: 10.7554/elife.74211] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 03/09/2022] [Indexed: 12/12/2022] Open
Abstract
Acute kidney injury is common, with ~13 million cases and 1.7 million deaths/year worldwide. A major cause is renal ischaemia, typically following cardiac surgery, renal transplant or severe haemorrhage. We examined the cause of the sustained reduction in renal blood flow ('no-reflow'), which exacerbates kidney injury even after an initial cause of compromised blood supply is removed. Adult male Sprague-Dawley rats, or NG2-dsRed male mice were used in this study. After 60 min kidney ischaemia and 30-60 min reperfusion, renal blood flow remained reduced, especially in the medulla, and kidney tubule damage was detected as Kim-1 expression. Constriction of the medullary descending vasa recta and cortical peritubular capillaries occurred near pericyte somata, and led to capillary blockages, yet glomerular arterioles and perfusion were unaffected, implying that the long-lasting decrease of renal blood flow contributing to kidney damage was generated by pericytes. Blocking Rho kinase to decrease pericyte contractility from the start of reperfusion increased the post-ischaemic diameter of the descending vasa recta capillaries at pericytes, reduced the percentage of capillaries that remained blocked, increased medullary blood flow and reduced kidney injury. Thus, post-ischaemic renal no-reflow, contributing to acute kidney injury, reflects pericytes constricting the descending vasa recta and peritubular capillaries. Pericytes are therefore an important therapeutic target for treating acute kidney injury.
Collapse
Affiliation(s)
- Felipe Freitas
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - David Attwell
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| |
Collapse
|
30
|
Zhang S, Liao XJ, Wang J, Shen Y, Shi HF, Zou Y, Ma CY, Wang XQ, Wang QG, Wang X, Xu MY, Cheng FF, Bai WZ. Temporal alterations in pericytes at the acute phase of ischemia/reperfusion in the mouse brain. Neural Regen Res 2022; 17:2247-2252. [PMID: 35259845 PMCID: PMC9083170 DOI: 10.4103/1673-5374.336876] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Pericytes, as the mural cells surrounding the microvasculature, play a critical role in the regulation of microcirculation; however, how these cells respond to ischemic stroke remains unclear. To determine the temporal alterations in pericytes after ischemia/reperfusion, we used the 1-hour middle cerebral artery occlusion model, which was examined at 2, 12, and 24 hours after reperfusion. Our results showed that in the reperfused regions, the cerebral blood flow decreased and the infarct volume increased with time. Furthermore, the pericytes in the infarct regions contracted and acted on the vascular endothelial cells within 24 hours after reperfusion. These effects may result in incomplete microcirculation reperfusion and a gradual worsening trend with time in the acute phase. These findings provide strong evidence for explaining the "no-reflow" phenomenon that occurs after recanalization in clinical practice.
Collapse
Affiliation(s)
- Shuang Zhang
- Beijing Key Laboratory, School of Basic Medical Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Xue-Jing Liao
- Beijing Key Laboratory, School of Basic Medical Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Jia Wang
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yi Shen
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Han-Fen Shi
- Beijing Key Laboratory, School of Basic Medical Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yan Zou
- Shineway Pharmaceutical Group Ltd., Shijiazhuang, Hebei Province, China
| | - Chong-Yang Ma
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Xue-Qian Wang
- Beijing Key Laboratory, School of Basic Medical Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Qing-Guo Wang
- Beijing Key Laboratory, School of Basic Medical Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Xu Wang
- Beijing Key Laboratory, School of Basic Medical Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Ming-Yang Xu
- Beijing Key Laboratory, School of Basic Medical Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Fa-Feng Cheng
- Beijing Key Laboratory, School of Basic Medical Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Wan-Zhu Bai
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
31
|
Del Franco AP, Chiang PP, Newman EA. Dilation of cortical capillaries is not related to astrocyte calcium signaling. Glia 2022; 70:508-521. [PMID: 34767261 PMCID: PMC8732319 DOI: 10.1002/glia.24119] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 10/12/2021] [Accepted: 10/31/2021] [Indexed: 12/30/2022]
Abstract
The brain requires an adequate supply of oxygen and nutrients to maintain proper function as neuronal activity varies. This is achieved, in part, through neurovascular coupling mechanisms that mediate local increases in blood flow through the dilation of arterioles and capillaries. The role of astrocytes in mediating this functional hyperemia response is controversial. Specifically, the function of astrocyte Ca2+ signaling is unclear. Cortical arterioles dilate in the absence of astrocyte Ca2+ signaling, but previous work suggests that Ca2+ increases are necessary for capillary dilation. This question has not been fully addressed in vivo, however, and we have reexamined the role of astrocyte Ca2+ signaling in vessel dilation in the barrel cortex of awake, behaving mice. We recorded evoked vessel dilations and astrocyte Ca2+ signaling in response to whisker stimulation. Experiments were carried out on WT and IP3R2 KO mice, a transgenic model where astrocyte Ca2+ signaling is substantially reduced. Compared to WT mice at rest, Ca2+ signaling in astrocyte endfeet contacting capillaries increased by 240% when whisker stimulation evoked running. In contrast, Ca2+ signaling was reduced to 9% of WT values in IP3R2 KO mice. In all three conditions, however, the amplitude of capillary dilation was largely unchanged. In addition, the latency to the onset of astrocyte Ca2+ signaling lagged behind dilation onset in most trials, although a subset of rapid onset Ca2+ events with latencies as short as 0.15 s occurred. In summary, we found that whisker stimulation-evoked capillary dilations occurred independent of astrocyte Ca2+ increases in the cerebral cortex.
Collapse
Affiliation(s)
- Armani P Del Franco
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, USA
| | - Pei-Pei Chiang
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, USA
| | - Eric A Newman
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
32
|
Pericyte dysfunction and loss of interpericyte tunneling nanotubes promote neurovascular deficits in glaucoma. Proc Natl Acad Sci U S A 2022; 119:2110329119. [PMID: 35135877 PMCID: PMC8851476 DOI: 10.1073/pnas.2110329119] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2021] [Indexed: 12/14/2022] Open
Abstract
Reduced blood flow and impaired neurovascular coupling are recognized features of glaucoma, the leading cause of irreversible blindness worldwide, but the mechanisms underlying these defects are unknown. Retinal pericytes regulate microcirculatory blood flow and coordinate neurovascular coupling through interpericyte tunneling nanotubes (IP-TNTs). Using two-photon microscope live imaging of the mouse retina, we found reduced capillary diameter and impaired blood flow at pericyte locations in eyes with high intraocular pressure, the most important risk factor to develop glaucoma. We show that IP-TNTs are structurally and functionally damaged by ocular hypertension, a response that disrupted light-evoked neurovascular coupling. Pericyte-specific inhibition of excessive Ca2+ influx rescued hemodynamic responses, protected IP-TNTs and neurovascular coupling, and enhanced retinal neuronal function as well as survival in glaucomatous retinas. Our study identifies pericytes and IP-TNTs as potential therapeutic targets to counter ocular pressure-related microvascular deficits, and provides preclinical proof of concept that strategies aimed to restore intrapericyte calcium homeostasis rescue autoregulatory blood flow and prevent neuronal dysfunction.
Collapse
|
33
|
Hartmann DA, Coelho-Santos V, Shih AY. Pericyte Control of Blood Flow Across Microvascular Zones in the Central Nervous System. Annu Rev Physiol 2022; 84:331-354. [PMID: 34672718 PMCID: PMC10480047 DOI: 10.1146/annurev-physiol-061121-040127] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The vast majority of the brain's vascular length is composed of capillaries, where our understanding of blood flow control remains incomplete. This review synthesizes current knowledge on the control of blood flow across microvascular zones by addressing issues with nomenclature and drawing on new developments from in vivo optical imaging and single-cell transcriptomics. Recent studies have highlighted important distinctions in mural cell morphology, gene expression, and contractile dynamics, which can explain observed differences in response to vasoactive mediators between arteriole, transitional, and capillary zones. Smooth muscle cells of arterioles and ensheathing pericytes of the arteriole-capillary transitional zone control large-scale, rapid changes in blood flow. In contrast, capillary pericytes downstream of the transitional zone act on slower and smaller scales and are involved in establishing resting capillary tone and flow heterogeneity. Many unresolved issues remain, including the vasoactive mediators that activate the different pericyte types in vivo, the role of pericyte-endothelial communication in conducting signals from capillaries to arterioles, and how neurological disease affects these mechanisms.
Collapse
Affiliation(s)
- David A Hartmann
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California, USA
| | - Vanessa Coelho-Santos
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington, USA;
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Andy Y Shih
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington, USA;
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
| |
Collapse
|
34
|
McConnell HL, Mishra A. Cells of the Blood-Brain Barrier: An Overview of the Neurovascular Unit in Health and Disease. Methods Mol Biol 2022; 2492:3-24. [PMID: 35733036 PMCID: PMC9987262 DOI: 10.1007/978-1-0716-2289-6_1] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
Abstract
The brain is endowed with highly specialized vasculature that is both structurally and functionally unique compared to vasculature supplying peripheral organs. The blood-brain barrier (BBB) is formed by endothelial cells of the cerebral vasculature and prevents extravasation of blood products into the brain to protect neural tissue and maintain a homeostatic environment. The BBB functions as part of the neurovascular unit (NVU), which is composed of neurons, astrocytes, and microglia in addition to the specialized endothelial cells, mural cells, and the basement membrane. Through coordinated intercellular signaling, these cells function as a dynamic unit to tightly regulate brain blood flow, vascular function, neuroimmune responses, and waste clearance. In this chapter, we review the functions of individual NVU components, describe neurovascular coupling as a classic example of NVU function, and discuss archetypal NVU pathophysiology during disease.
Collapse
Affiliation(s)
- Heather L McConnell
- Department of Neurology, Jungers Center for Neurosciences Research, Oregon Health & Science University, Portland, OR, USA
- Office of Academic Development, Houston Methodist Research Institute, Houston, TX, USA
| | - Anusha Mishra
- Department of Neurology, Jungers Center for Neurosciences Research, Oregon Health & Science University, Portland, OR, USA.
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
35
|
Butsabong T, Felippe M, Campagnolo P, Maringer K. The emerging role of perivascular cells (pericytes) in viral pathogenesis. J Gen Virol 2021; 102. [PMID: 34424156 PMCID: PMC8513640 DOI: 10.1099/jgv.0.001634] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Viruses may exploit the cardiovascular system to facilitate transmission or within-host dissemination, and the symptoms of many viral diseases stem at least in part from a loss of vascular integrity. The microvascular architecture is comprised of an endothelial cell barrier ensheathed by perivascular cells (pericytes). Pericytes are antigen-presenting cells (APCs) and play crucial roles in angiogenesis and the maintenance of microvascular integrity through complex reciprocal contact-mediated and paracrine crosstalk with endothelial cells. We here review the emerging ways that viruses interact with pericytes and pay consideration to how these interactions influence microvascular function and viral pathogenesis. Major outcomes of virus-pericyte interactions include vascular leakage or haemorrhage, organ tropism facilitated by barrier disruption, including viral penetration of the blood-brain barrier and placenta, as well as inflammatory, neurological, cognitive and developmental sequelae. The underlying pathogenic mechanisms may include direct infection of pericytes, pericyte modulation by secreted viral gene products and/or the dysregulation of paracrine signalling from or to pericytes. Viruses we cover include the herpesvirus human cytomegalovirus (HCMV, Human betaherpesvirus 5), the retrovirus human immunodeficiency virus (HIV; causative agent of acquired immunodeficiency syndrome, AIDS, and HIV-associated neurocognitive disorder, HAND), the flaviviruses dengue virus (DENV), Japanese encephalitis virus (JEV) and Zika virus (ZIKV), and the coronavirus severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2; causative agent of coronavirus disease 2019, COVID-19). We touch on promising pericyte-focussed therapies for treating the diseases caused by these important human pathogens, many of which are emerging viruses or are causing new or long-standing global pandemics.
Collapse
Affiliation(s)
- Teemapron Butsabong
- Department of Biochemical Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH, UK
| | - Mariana Felippe
- Department of Biochemical Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH, UK
| | - Paola Campagnolo
- Department of Biochemical Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH, UK
| | - Kevin Maringer
- The Pirbright Institute, Pirbright, Surrey, GU24 0NF, UK
| |
Collapse
|
36
|
Cao L, Zhou Y, Chen M, Li L, Zhang W. Pericytes for Therapeutic Approaches to Ischemic Stroke. Front Neurosci 2021; 15:629297. [PMID: 34239409 PMCID: PMC8259582 DOI: 10.3389/fnins.2021.629297] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 05/11/2021] [Indexed: 12/18/2022] Open
Abstract
Pericytes are perivascular multipotent cells located on capillaries. Although pericytes are discovered in the nineteenth century, recent studies have found that pericytes play an important role in maintaining the blood—brain barrier (BBB) and regulating the neurovascular system. In the neurovascular unit, pericytes perform their functions by coordinating the crosstalk between endothelial, glial, and neuronal cells. Dysfunction of pericytes can lead to a variety of diseases, including stroke and other neurological disorders. Recent studies have suggested that pericytes can serve as a therapeutic target in ischemic stroke. In this review, we first summarize the biology and functions of pericytes in the central nervous system. Then, we focus on the role of dysfunctional pericytes in the pathogenesis of ischemic stroke. Finally, we discuss new therapies for ischemic stroke based on targeting pericytes.
Collapse
Affiliation(s)
- Lu Cao
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanbo Zhou
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mengguang Chen
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Li Li
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Wei Zhang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
37
|
Affiliation(s)
- Adam Institoris
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Grant R Gordon
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
38
|
Iturriaga-Goyon E, Buentello-Volante B, Magaña-Guerrero FS, Garfias Y. Future Perspectives of Therapeutic, Diagnostic and Prognostic Aptamers in Eye Pathological Angiogenesis. Cells 2021; 10:cells10061455. [PMID: 34200613 PMCID: PMC8227682 DOI: 10.3390/cells10061455] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/04/2021] [Accepted: 06/05/2021] [Indexed: 12/23/2022] Open
Abstract
Aptamers are single-stranded DNA or RNA oligonucleotides that are currently used in clinical trials due to their selectivity and specificity to bind small molecules such as proteins, peptides, viral particles, vitamins, metal ions and even whole cells. Aptamers are highly specific to their targets, they are smaller than antibodies and fragment antibodies, they can be easily conjugated to multiple surfaces and ions and controllable post-production modifications can be performed. Aptamers have been therapeutically used for age-related macular degeneration, cancer, thrombosis and inflammatory diseases. The aim of this review is to highlight the therapeutic, diagnostic and prognostic possibilities associated with aptamers, focusing on eye pathological angiogenesis.
Collapse
Affiliation(s)
- Emilio Iturriaga-Goyon
- MD/PhD (PECEM) Program, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
- Cell and Tissue Biology, Research Unit, Institute of Ophthalmology, Conde de Valenciana, Chimalpopoca 14, Mexico City 06800, Mexico; (B.B.-V.); (F.S.M.-G.)
- Department of Biochemistry, Facultad de Medicina, Universidad Nacional Autónoma de México, Av. Universidad 3000, Mexico City 04510, Mexico
| | - Beatriz Buentello-Volante
- Cell and Tissue Biology, Research Unit, Institute of Ophthalmology, Conde de Valenciana, Chimalpopoca 14, Mexico City 06800, Mexico; (B.B.-V.); (F.S.M.-G.)
| | - Fátima Sofía Magaña-Guerrero
- Cell and Tissue Biology, Research Unit, Institute of Ophthalmology, Conde de Valenciana, Chimalpopoca 14, Mexico City 06800, Mexico; (B.B.-V.); (F.S.M.-G.)
| | - Yonathan Garfias
- Cell and Tissue Biology, Research Unit, Institute of Ophthalmology, Conde de Valenciana, Chimalpopoca 14, Mexico City 06800, Mexico; (B.B.-V.); (F.S.M.-G.)
- Department of Biochemistry, Facultad de Medicina, Universidad Nacional Autónoma de México, Av. Universidad 3000, Mexico City 04510, Mexico
- Correspondence:
| |
Collapse
|
39
|
Brain capillary pericytes exert a substantial but slow influence on blood flow. Nat Neurosci 2021; 24:633-645. [PMID: 33603231 PMCID: PMC8102366 DOI: 10.1038/s41593-020-00793-2] [Citation(s) in RCA: 219] [Impact Index Per Article: 54.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 12/23/2020] [Indexed: 01/30/2023]
Abstract
The majority of the brain's vasculature is composed of intricate capillary networks lined by capillary pericytes. However, it remains unclear whether capillary pericytes influence blood flow. Using two-photon microscopy to observe and manipulate brain capillary pericytes in vivo, we find that their optogenetic stimulation decreases lumen diameter and blood flow, but with slower kinetics than similar stimulation of mural cells on upstream pial and precapillary arterioles. This slow vasoconstriction was inhibited by the clinically used vasodilator fasudil, a Rho-kinase inhibitor that blocks contractile machinery. Capillary pericytes were also slower to constrict back to baseline following hypercapnia-induced dilation, and slower to dilate towards baseline following optogenetically induced vasoconstriction. Optical ablation of single capillary pericytes led to sustained local dilation and a doubling of blood cell flux selectively in capillaries lacking pericyte contact. These data indicate that capillary pericytes contribute to basal blood flow resistance and slow modulation of blood flow throughout the brain.
Collapse
|
40
|
Alarcon-Martinez L, Yemisci M, Dalkara T. Pericyte morphology and function. Histol Histopathol 2021; 36:633-643. [PMID: 33595091 DOI: 10.14670/hh-18-314] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The proper delivery of blood is essential for healthy neuronal function. The anatomical substrate for this precise mechanism is the neurovascular unit, which is formed by neurons, glial cells, endothelia, smooth muscle cells, and pericytes. Based on their particular location on the vessel wall, morphology, and protein expression, pericytes have been proposed as cells capable of regulating capillary blood flow. Pericytes are located around the microvessels, wrapping them with their processes. Their morphology and protein expression substantially vary along the vascular tree. Their contractibility is mediated by a unique cytoskeleton organization formed by filaments of actin that allows pericyte deformability with the consequent mechanical force transferred to the extracellular matrix for changing the diameter. Pericyte ultrastructure is characterized by large mitochondria likely to provide energy to regulate intracellular calcium concentration and fuel contraction. Accordingly, pericytes with compromised energy show a sustained intracellular calcium increase that leads to persistent microvascular constriction. Pericyte morphology is highly plastic and adapted for varying contractile capability along the microvascular tree, making pericytes ideal cells to regulate the capillary blood flow in response to local neuronal activity. Besides the vascular regulation, pericytes also play a role in the maintenance of the blood-brain/retina barrier, neovascularization and angiogenesis, and leukocyte transmigration. Here, we review the morphological and functional features of the pericytes as well as potential specific markers for the study of pericytes in the brain and retina.
Collapse
Affiliation(s)
- Luis Alarcon-Martinez
- Department of Neuroscience and Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Université de Montréal, Montréal, QC, Canada.
| | - Muge Yemisci
- Department of Neurology, Faculty of Medicine, Hacettepe University, Ankara, Turkey.,Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| | - Turgay Dalkara
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| |
Collapse
|
41
|
Kaul S, Methner C, Mishra A. The role of pericytes in hyperemia-induced capillary de-recruitment following stenosis. ACTA ACUST UNITED AC 2020; 1:163-169. [PMID: 33778770 DOI: 10.1007/s43152-020-00017-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Purpose The microvascular capillary network is ensheathed by cells called pericytes - a heterogeneous population of mural cells derived from multiple lineages. Pericytes play a multifaceted role in the body, including in vascular structure and permeability, regulation of local blood flow, immune and wound healing functions, induction of angiogenesis, and generation of various progenitor cells. Here, we consider the role of pericytes in capillary de-recruitment, a pathophysiologic phenomenon that is observed following hyperemic stimuli in the presence of a stenosis and attenuates the hyperemic response. Recent Findings We discuss recent observations that conclusively demonstrate pericytes to be the cellular structures that contract in response to hyperemic stimuli when an upstream arterial stenosis is present. This response constricts capillaries, which is likely aimed at maintaining capillary hydrostatic pressure, an important factor in tissue homeostasis. Nonetheless, the ensuing attenuation of the hyperemic response can lead to a decrease in energy supply and negatively impact tissue health. Summary Therapeutics aimed at preventing pericyte-mediated capillary de-recruitment may prove beneficial in conditions such as coronary stenosis and peripheral arterial disease by reducing restriction in hyperemic flow. Identification of the pericyte subtypes involved in this de-recruitment and the underlying molecular mechanisms regulating this process will greatly assist this purpose.
Collapse
Affiliation(s)
- Sanjiv Kaul
- Knight Cardiovascular Institute Oregon Health & Science University, Portland, Oregon, USA
| | - Carmen Methner
- Knight Cardiovascular Institute Oregon Health & Science University, Portland, Oregon, USA
| | - Anusha Mishra
- Knight Cardiovascular Institute Oregon Health & Science University, Portland, Oregon, USA.,Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
42
|
Mantegazza A, Ungari M, Clavica F, Obrist D. Local vs. Global Blood Flow Modulation in Artificial Microvascular Networks: Effects on Red Blood Cell Distribution and Partitioning. Front Physiol 2020; 11:566273. [PMID: 33123027 PMCID: PMC7571285 DOI: 10.3389/fphys.2020.566273] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 08/12/2020] [Indexed: 12/12/2022] Open
Abstract
Our understanding of cerebral blood flow (CBF) regulation during functional activation is still limited. Alongside with the accepted role of smooth muscle cells in controlling the arteriolar diameter, a new hypothesis has been recently formulated suggesting that CBF may be modulated by capillary diameter changes mediated by pericytes. In this study, we developed in vitro microvascular network models featuring a valve enabling the dilation of a specific micro-channel. This allowed us to investigate the non-uniform red blood cell (RBC) partitioning at microvascular bifurcations (phase separation) and the hematocrit distribution at rest and for two scenarios modeling capillary and arteriolar dilation. RBC partitioning showed similar phase separation behavior during baseline and activation. Results indicated that the RBCs at diverging bifurcations generally enter the high-flow branch (classical partitioning). Inverse behavior (reverse partitioning) was observed for skewed hematocrit profiles in the parent vessel of bifurcations, especially for high RBC velocity (i.e., arteriolar activation). Moreover, results revealed that a local capillary dilation, as it may be mediated in vivo by pericytes, led to a localized increase of RBC flow and a heterogeneous hematocrit redistribution within the whole network. In case of a global increase of the blood flow, as it may be achieved by dilating an arteriole, a homogeneous increase of RBC flow was observed in the whole network and the RBCs were concentrated along preferential pathways. In conclusion, overall increase of RBC flow could be obtained by arteriolar and capillary dilation, but only capillary dilation was found to alter the perfusion locally and heterogeneously.
Collapse
Affiliation(s)
- Alberto Mantegazza
- ARTORG Center for Biomedical Engineering Research, University of Bern, Bern, Switzerland
| | - Matteo Ungari
- ARTORG Center for Biomedical Engineering Research, University of Bern, Bern, Switzerland
| | - Francesco Clavica
- ARTORG Center for Biomedical Engineering Research, University of Bern, Bern, Switzerland.,Integrated Actuators Laboratory, École Polytechnique Fédérale de Lausanne, Neuchâtel, Switzerland
| | - Dominik Obrist
- ARTORG Center for Biomedical Engineering Research, University of Bern, Bern, Switzerland
| |
Collapse
|
43
|
Alarcon-Martinez L, Villafranca-Baughman D, Quintero H, Kacerovsky JB, Dotigny F, Murai KK, Prat A, Drapeau P, Di Polo A. Interpericyte tunnelling nanotubes regulate neurovascular coupling. Nature 2020; 585:91-95. [DOI: 10.1038/s41586-020-2589-x] [Citation(s) in RCA: 179] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 07/06/2020] [Indexed: 12/13/2022]
|
44
|
Wareham LK, Calkins DJ. The Neurovascular Unit in Glaucomatous Neurodegeneration. Front Cell Dev Biol 2020; 8:452. [PMID: 32656207 PMCID: PMC7325980 DOI: 10.3389/fcell.2020.00452] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 05/15/2020] [Indexed: 12/31/2022] Open
Abstract
Glaucoma is a neurodegenerative disease of the visual system and leading cause of blindness worldwide. The disease is associated with sensitivity to intraocular pressure (IOP), which over a large range of magnitudes stresses retinal ganglion cell (RGC) axons as they pass through the optic nerve head in forming the optic projection to the brain. Despite clinical efforts to lower IOP, which is the only modifiable risk factor for glaucoma, RGC degeneration and ensuing loss of vision often persist. A major contributor to failure of hypotensive regimens is the multifactorial nature of how IOP-dependent stress influences RGC physiology and structure. This stress is conveyed to the RGC axon through interactions with structural, glial, and vascular components in the nerve head and retina. These interactions promote pro-degenerative pathways involving biomechanical, metabolic, oxidative, inflammatory, immunological and vascular challenges to the microenvironment of the ganglion cell and its axon. Here, we focus on the contribution of vascular dysfunction and breakdown of neurovascular coupling in glaucoma. The vascular networks of the retina and optic nerve head have evolved complex mechanisms that help to maintain a continuous blood flow and supply of metabolites despite fluctuations in ocular perfusion pressure. In healthy tissue, autoregulation and neurovascular coupling enable blood flow to stay tightly controlled. In glaucoma patients evidence suggests these pathways are dysfunctional, thus highlighting a potential role for pathways involved in vascular dysfunction in progression and as targets for novel therapeutic intervention.
Collapse
Affiliation(s)
- Lauren K Wareham
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN, United States
| | - David J Calkins
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
45
|
Nelson AR, Sagare MA, Wang Y, Kisler K, Zhao Z, Zlokovic BV. Channelrhodopsin Excitation Contracts Brain Pericytes and Reduces Blood Flow in the Aging Mouse Brain in vivo. Front Aging Neurosci 2020; 12:108. [PMID: 32410982 PMCID: PMC7201096 DOI: 10.3389/fnagi.2020.00108] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 03/30/2020] [Indexed: 01/02/2023] Open
Abstract
Brains depend on blood flow for the delivery of oxygen and nutrients essential for proper neuronal and synaptic functioning. French physiologist Rouget was the first to describe pericytes in 1873 as regularly arranged longitudinal amoeboid cells on capillaries that have a muscular coat, implying that these are contractile cells that regulate blood flow. Although there have been >30 publications from different groups, including our group, demonstrating that pericytes are contractile cells that can regulate hemodynamic responses in the brain, the role of pericytes in controlling cerebral blood flow (CBF) has not been confirmed by all studies. Moreover, recent studies using different optogenetic models to express light-sensitive channelrhodopsin-2 (ChR2) cation channels in pericytes were not conclusive; one, suggesting that pericytes expressing ChR2 do not contract after light stimulus, and the other, demonstrating contraction of pericytes expressing ChR2 after light stimulus. Since two-photon optogenetics provides a powerful tool to study mechanisms of blood flow regulation at the level of brain capillaries, we re-examined the contractility of brain pericytes in vivo using a new optogenetic model developed by crossing our new inducible pericyte-specific CreER mouse line with ChR2 mice. We induced expression of ChR2 in pericytes with tamoxifen, excited ChR2 by 488 nm light, and monitored pericyte contractility, brain capillary diameter changes, and red blood cell (RBC) velocity in aged mice by in vivo two-photon microscopy. Excitation of ChR2 resulted in pericyte contraction followed by constriction of the underlying capillary leading to approximately an 8% decrease (p = 0.006) in capillary diameter. ChR2 excitation in pericytes substantially reduced capillary RBC flow by 42% (p = 0.03) during the stimulation period compared to the velocity before stimulation. Our data suggests that pericytes contract in vivo and regulate capillary blood flow in the aging mouse brain. By extension, this might have implications for neurological disorders of the aging human brain associated with neurovascular dysfunction and pericyte loss such as stroke and Alzheimer's disease.
Collapse
Affiliation(s)
| | | | | | | | | | - Berislav V. Zlokovic
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
46
|
Wang D, Cheng X, Fang H, Ren Y, Li X, Ren W, Xue B, Yang C. Effect of cold stress on ovarian & uterine microcirculation in rats and the role of endothelin system. Reprod Biol Endocrinol 2020; 18:29. [PMID: 32290862 PMCID: PMC7155299 DOI: 10.1186/s12958-020-00584-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 03/27/2020] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Cold, an environmental factor, induces many reproductive diseases. It is known that endothelin (ET) is a potent vasoconstrictor, and cold stress can increase the expression of ET and its receptors. The cold stress rat model was developed to examine two parameters: (1) the effects of cold stress on ovarian and uterine morphology, function, and microvascular circulation and (2) possible mechanisms of ET and its receptors involved in cold stress-induced menstruation disorders. METHODS The rat cold stress model was prepared with an ice water bath. The estrous cycle was observed by methylene blue and hematoxylin and eosin (H&E) staining. Serum estradiol 2 (E2), testosterone (T), progesterone (P) were detected by radioimmunoassay. Hemorheology indices were measured. The real-time blood flow of auricle and uterine surfaces was measured. Expressions of CD34 and α-SMA in ovarian and uterine tissues were detected by immunohistochemistry. ET-1 contents in serum were tested, and expressions of ET-receptor types A and B (ET-AR and ET-BR) in ovarian tissues were detected via Western blotting. RESULTS Cold stress extended the estrous cycle, thereby causing reproductive hormone disorder, imbalance of local endothelin/nitric oxide expression, and microcirculation disturbance. Cold-stress led to up-regulation of ET-AR expression and protein and down-regulation of ET-BR expression in rats. CONCLUSIONS This study suggests that the reason for cold stress-induced dysfunction in reproductive organs may be closely related to the imbalance of ET-1 and its receptor expressions, leading to microvascular circulation disorders in local tissues.
Collapse
Affiliation(s)
- Di Wang
- grid.488206.00000 0004 4912 1751Hebei University of Chinese Medicine, No.326, Xinshi South Road, Qiaoxi District, Shijiazhuang, 050091 Hebei Province China
| | - Xiumei Cheng
- grid.488206.00000 0004 4912 1751Hebei University of Chinese Medicine, No.326, Xinshi South Road, Qiaoxi District, Shijiazhuang, 050091 Hebei Province China
| | - Huimin Fang
- grid.488206.00000 0004 4912 1751Hebei University of Chinese Medicine, No.326, Xinshi South Road, Qiaoxi District, Shijiazhuang, 050091 Hebei Province China
| | - Yanqing Ren
- grid.488206.00000 0004 4912 1751Hebei University of Chinese Medicine, No.326, Xinshi South Road, Qiaoxi District, Shijiazhuang, 050091 Hebei Province China
| | - Xinhua Li
- grid.488206.00000 0004 4912 1751Hebei University of Chinese Medicine, No.326, Xinshi South Road, Qiaoxi District, Shijiazhuang, 050091 Hebei Province China
| | - Weiwei Ren
- grid.488206.00000 0004 4912 1751Hebei University of Chinese Medicine, No.326, Xinshi South Road, Qiaoxi District, Shijiazhuang, 050091 Hebei Province China
| | - Bing Xue
- grid.488206.00000 0004 4912 1751Hebei University of Chinese Medicine, No.326, Xinshi South Road, Qiaoxi District, Shijiazhuang, 050091 Hebei Province China
| | - Cairui Yang
- grid.488206.00000 0004 4912 1751Hebei University of Chinese Medicine, No.326, Xinshi South Road, Qiaoxi District, Shijiazhuang, 050091 Hebei Province China
| |
Collapse
|
47
|
Alpha-Smooth Muscle Actin-Positive Perivascular Cells in Diabetic Retina and Choroid. Int J Mol Sci 2020; 21:ijms21062158. [PMID: 32245120 PMCID: PMC7139401 DOI: 10.3390/ijms21062158] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/18/2020] [Accepted: 03/18/2020] [Indexed: 02/07/2023] Open
Abstract
Structural alterations of pericytes in microvessels are important features of diabetic retinopathy. Although capillary pericytes had been known not to have α-smooth muscle actin (αSMA), a recent study revealed that a specific fixation method enabled the visualization of αSMA along retinal capillaries. In this study, we applied snap-fixation in wild type and streptozotocin-induced diabetic mice to evaluate the differences in vascular smooth muscle cells of the retina and the choroid. Mice eyeballs were fixed in ice-cold methanol to prevent the depolymerization of filamentous actin. Snap-fixated retina showed αSMA expression in higher-order branches along the capillaries as well as the arterioles and venules, which were not detected by paraformaldehyde fixation. In contrast, most choriocapillaris, except those close to the arterioles, were not covered with αSMA-positive perivascular mural cells. Large choroidal vessels were covered with more αSMA-positive cells in the snap-fixated eyes. Diabetes induced less coverage of αSMA-positive perivascular mural cells overall, but they reached higher-order branches of the retinal capillaries, which was prominent in the aged mice. More αSMA-positive pericytes were observed in the choroid of diabetic mice, but the αSMA-positive expression reduced with aging. This study suggests the potential role of smooth muscle cells in the pathogenesis of age-related diabetic retinopathy and choroidopathy.
Collapse
|
48
|
Nwadozi E, Rudnicki M, Haas TL. Metabolic Coordination of Pericyte Phenotypes: Therapeutic Implications. Front Cell Dev Biol 2020; 8:77. [PMID: 32117997 PMCID: PMC7033550 DOI: 10.3389/fcell.2020.00077] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 01/29/2020] [Indexed: 12/15/2022] Open
Abstract
Pericytes are mural vascular cells found predominantly on the abluminal wall of capillaries, where they contribute to the maintenance of capillary structural integrity and vascular permeability. Generally quiescent cells in the adult, pericyte activation and proliferation occur during both physiological and pathological vascular and tissue remodeling. A considerable body of research indicates that pericytes possess attributes of a multipotent adult stem cell, as they are capable of self-renewal as well as commitment and differentiation into multiple lineages. However, pericytes also display phenotypic heterogeneity and recent studies indicate that lineage potential differs between pericyte subpopulations. While numerous microenvironmental cues and cell signaling pathways are known to regulate pericyte functions, the roles that metabolic pathways play in pericyte quiescence, self-renewal or differentiation have been given limited consideration to date. This review will summarize existing data regarding pericyte metabolism and will discuss the coupling of signal pathways to shifts in metabolic pathway preferences that ultimately regulate pericyte quiescence, self-renewal and trans-differentiation. The association between dysregulated metabolic processes and development of pericyte pathologies will be highlighted. Despite ongoing debate regarding pericyte classification and their functional capacity for trans-differentiation in vivo, pericytes are increasingly exploited as a cell therapy tool to promote tissue healing and regeneration. Ultimately, the efficacy of therapeutic approaches hinges on the capacity to effectively control/optimize the fate of the implanted pericytes. Thus, we will identify knowledge gaps that need to be addressed to more effectively harness the opportunity for therapeutic manipulation of pericytes to control pathological outcomes in tissue remodeling.
Collapse
Affiliation(s)
| | | | - Tara L. Haas
- School of Kinesiology and Health Science, Angiogenesis Research Group and Muscle Health Research Centre, York University, Toronto, ON, Canada
| |
Collapse
|