1
|
Ghosh S, Bishnoi B, Das S. Artery regeneration: Molecules, mechanisms and impact on organ function. Semin Cell Dev Biol 2025; 171:103611. [PMID: 40318557 DOI: 10.1016/j.semcdb.2025.103611] [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: 10/07/2024] [Revised: 02/24/2025] [Accepted: 03/25/2025] [Indexed: 05/07/2025]
Abstract
Replenishment of artery cells to repair or create new arteries is a promising strategy to re-vascularize ischemic tissue. However, limited understanding of cellular and molecular programs associated with artery (re-)growth impedes our efforts towards designing optimal therapeutic approaches. In this review, we summarize different cellular mechanisms that drive injury-induced artery regeneration in distinct organs and organisms. Artery formation during embryogenesis includes migration, self-amplification, and changes in cell fates. These processes are coordinated by multiple signaling pathways, like Vegf, Wnt, Notch, Cxcr4; many of which, also involved in injury-induced vascular responses. We also highlight how physiological and environmental factors determine the extent of arterial re-vascularization. Finally, we discuss different in vitro cellular reprogramming and tissue engineering approaches to promote artery regeneration, in vivo. This review provides the current understanding of endothelial cell fate reprogramming and explores avenues for regenerating arteries to restore organ function through efficient revascularization.
Collapse
Affiliation(s)
- Swarnadip Ghosh
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, KA 560065, India
| | - Bhavnesh Bishnoi
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, KA 560065, India
| | - Soumyashree Das
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, KA 560065, India.
| |
Collapse
|
2
|
Bisen S, Gogoi P, Sharma A, Mukhopadhyay CS, Singh NK. A Disintegrin and Metalloproteinase 10 Regulates Ephrin B2-Mediated Endothelial Cell Sprouting and Ischemic Retinopathy. THE AMERICAN JOURNAL OF PATHOLOGY 2025:S0002-9440(25)00114-2. [PMID: 40252970 DOI: 10.1016/j.ajpath.2025.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 03/14/2025] [Accepted: 03/21/2025] [Indexed: 04/21/2025]
Abstract
Retinal neovascularization is the leading cause of visual impairment in diabetic retinopathy, retinopathy of prematurity, and age-related macular degeneration. The extracellular matrix breakdown by metalloproteinase leads to vascular complications in various proliferative retinopathies. A disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) is involved in physiological angiogenesis. However, limited information exists regarding the role of ADAM10 in proliferative retinopathies. In this study, the levels of active ADAM10 were significantly up-regulated in the ischemic retina, and down-regulation or inactivation of ADAM10 significantly inhibited the proliferation, sprouting, migration, and tube formation of human retinal microvascular endothelial cell. Furthermore, the endothelial cell (EC)-specific deletion of ADAM10 (ADAM10iΔEC) significantly attenuated vascular leakage, edema, endothelial cell sprouting, and retinal neovascularization in ischemic retinas of mice exposed to oxygen-induced retinopathy. In experiments investigating the mechanisms through which ADAM10 regulated pathologic angiogenesis, ADAM10 regulated ephrin B2 (EfnB2) expression in endothelial cells. Down-regulation of EfnB2 expression influenced human retinal microvascular endothelial cell proliferation, migration, sprouting, and tube formation. In addition, a significant up-regulation of EfnB2 expression in the ischemic retina was detected. EC-specific depletion of ADAM10 significantly reduced EfnB2 expression, suggesting its involvement in ADAM10-regulated retinal neovascularization. The findings demonstrate how EC-specific ADAM10 regulates pathologic retinal neovascularization in the ischemic retina, indicating its significance as a potential therapeutic target for proliferative retinopathies.
Collapse
Affiliation(s)
- Shivantika Bisen
- Integrative Biosciences Center, Wayne State University, Detroit, Michigan; Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, Michigan
| | - Purnima Gogoi
- Integrative Biosciences Center, Wayne State University, Detroit, Michigan; Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, Michigan; Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Anamika Sharma
- Integrative Biosciences Center, Wayne State University, Detroit, Michigan; Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, Michigan
| | - Chandra S Mukhopadhyay
- Department of Bioinformatics, College of Animal Biotechnology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, India
| | - Nikhlesh K Singh
- Integrative Biosciences Center, Wayne State University, Detroit, Michigan; Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, Michigan.
| |
Collapse
|
3
|
Faber JE. Collateral blood vessels in stroke and ischemic disease: Formation, physiology, rarefaction, remodeling. J Cereb Blood Flow Metab 2025:271678X251322378. [PMID: 40072222 PMCID: PMC11904929 DOI: 10.1177/0271678x251322378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
Collateral blood vessels are unique, naturally occurring endogenous bypass vessels that provide alternative pathways for oxygen delivery in obstructive arterial conditions and diseases. Surprisingly however, the capacity of the collateral circulation to provide protection varies greatly among individuals, resulting in a significant fraction having poor collateral circulation in their tissues. We recently reviewed evidence that the presence of naturally-occurring polymorphisms in genes that determine the number and diameter of collaterals that form during development (ie, genetic background), is a major contributor to this variation. The purpose of this review is to summarize current understanding of the other determinants of collateral blood flow, drawing on both animal and human studies. These include the level of smooth muscle tone in collaterals, hemodynamic forces, how collaterals form during development (collaterogenesis), de novo formation of additional new collaterals during adulthood, loss of collaterals with aging and cardiovascular risk factor presence (rarefaction), and collateral remodeling (structural lumen enlargement). We also review emerging evidence that collaterals not only provide protection in ischemic conditions but may also serve a physiological function in healthy individuals. Primary focus is on studies conducted in brain, however relevant findings in other tissues are also reviewed, as are questions for future investigation.
Collapse
Affiliation(s)
- James E Faber
- Department of Cell Biology and Physiology, Curriculum in Neuroscience, McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
4
|
Lin KC, He W, Wang D, Yao ML, Chen J, Chen MF, Zhang GG, Li CC, Zhu LP, Bai YP. Selumetinib promotes coronary collateral circulation by inducing M2-like macrophage polarization following myocardial infarction. Acta Pharmacol Sin 2025:10.1038/s41401-025-01508-8. [PMID: 40055526 DOI: 10.1038/s41401-025-01508-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 02/10/2025] [Indexed: 03/12/2025]
Abstract
Coronary collateral circulation (CCC) construction could be a practical therapeutic strategy for patients following myocardial infarction (MI), yet effective therapeutic drugs remain scarce. In this study we conducted database federation analyses to identify FDA-approved drugs that could promote CCC after MI injury. By comparing the differentially expressed genes in peripheral blood mononuclear cells (PBMCs) from two public gene profiles: one comparing patients with good versus poor CCC, and another with good versus poor heart function, the overlapped genes were analyzed using CMap, a popular resource designed for FDA approved drug. As a result, selumetinib emerged as a potential therapeutic drug to facilitate CCC formation. In MI mouse model induced by permanent ligation of left anterior descending (LAD) coronary artery, administration of selumetinib (2.5 mg/kg, i.p.) at the indicated time-points significantly enhanced CCC by promoting the polarization of macrophages from the pro-inflammatory M1-like phenotype to the pro-angiogenic M2-like phenotype, which was confirmed by 3D visualization through micro-CT imaging and immunofluorescent staining. We demonstrated that selumetinib (5 μM) promoted THP-1 differentiated into M2-like phenotype in vitro, and increased VEGFA secretion. Selumetinib-treated macrophages significantly enhanced in vitro angiogenesis of HUVECs in cocultured assay. We found that selumetinib (2.5 and 5 μM) dose-dependently inhibited the expression of the RIT1 in THP-1 derived M1 macrophage; knockdown of RIT1 significantly polarized M2-like phenotype via the MAPK/ERK1/2 signaling pathway, which was equal to the efficiency of selumetinib. In rescued experiments, specific overexpression of RIT1 in macrophage by injecting with targeting F4/80 promoter AAV9 in mice, could block the M2-like phenotype shifts and CCC formation by selumetinib. Finally, honokiol, a MAPK/ERK1/2 agonist was able to reverse the effects of selumetinib on CCC in mice with MI. In conclusion, selumetinib possesses therapeutic potential for induction of CCC formation after MI.
Collapse
Affiliation(s)
- Ke-Chuan Lin
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Wei He
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Dan Wang
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Mei-Lian Yao
- Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jing Chen
- Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Mei-Fang Chen
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Guo-Gang Zhang
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Chuan-Chang Li
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Ling-Ping Zhu
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Yong-Ping Bai
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Department of Cardiology, The Xiangya Hospital, Central South University, Changsha, 410013, China.
| |
Collapse
|
5
|
Faber JE. Genetic determinants of insufficiency of the collateral circulation. J Cereb Blood Flow Metab 2025:271678X251317880. [PMID: 39901795 DOI: 10.1177/0271678x251317880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2025]
Abstract
It has been estimated that approximately two million neurons, sixteen billion synapses and twelve kilometers of axons are lost each minute following anterior large-vessel stroke. The level of collateral blood flow has become recognized as a primary determinant of the pace of this loss and an important factor in clinical decision-making. Many of the topics in this review cover recent developments that have not been reviewed elsewhere. These include that: the number and diameter of collaterals and collateral blood flow vary greatly in the brain and other tissues of healthy individuals; a large percentage of individuals are deficient in collaterals; the underlying mechanism arises primarily from naturally occurring polymorphisms in genes/genetic loci within the pathway that drives collateral formation during development; evidence indicates collateral abundance does not exhibit sexual dimorphism; and that collaterals-besides their function as endogenous bypass vessels-may have a physiological role in optimizing oxygen delivery. Animal and human studies in brain and other tissues, where available, are reviewed. Details of many of the studies are provided so that the strength of the findings and conclusions can be assessed without consulting the original literature. Key questions that remain unanswered and strategies to address them are also discussed.
Collapse
Affiliation(s)
- James E Faber
- Department of Cell Biology and Physiology, Curriculum in Neuroscience, McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
6
|
Sinha A, Gupta M, Bhaskar SMM. Evolucollateral dynamics in stroke: Evolutionary pathophysiology, remodelling and emerging therapeutic strategies. Eur J Neurosci 2024; 60:6779-6798. [PMID: 39498733 DOI: 10.1111/ejn.16585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 10/13/2024] [Accepted: 10/15/2024] [Indexed: 11/07/2024]
Abstract
Leptomeningeal collaterals (LMCs) are crucial in mitigating the impact of acute ischemic stroke (AIS) by providing alternate blood flow routes when primary arteries are obstructed. This article explores the evolutionary pathophysiology of LMCs, highlighting their critical function in stroke and the genetic and molecular mechanisms governing their development and remodelling. We address the translational challenges of applying animal model findings to human clinical scenarios, emphasizing the need for further research to validate emerging therapies-such as pharmacological agents, gene therapy and mechanical interventions-in clinical settings, aimed at enhancing collateral perfusion. Computational modelling emerges as a promising method for integrating experimental data, which requires precise parameterization and empirical validation. We introduce the 'Evolucollateral Dynamics' hypothesis, proposing a novel framework that incorporates evolutionary biology principles into therapeutic strategies, offering new perspectives on enhancing collateral circulation. This hypothesis emphasizes the role of genetic predispositions and environmental influences on collateral circulation, which may impact therapeutic strategies and optimize treatment outcomes. Future research must incorporate human clinical data to create robust treatment protocols, thereby maximizing the therapeutic potential of LMCs and improving outcomes for stroke patients.
Collapse
Affiliation(s)
- Akansha Sinha
- Global Health Neurology Lab, Sydney, NSW, Australia
- UNSW Medicine and Health, University of New South Wales (UNSW), South West Sydney Clinical Campuses, Sydney, NSW, Australia
| | - Muskaan Gupta
- Global Health Neurology Lab, Sydney, NSW, Australia
- UNSW Medicine and Health, University of New South Wales (UNSW), South West Sydney Clinical Campuses, Sydney, NSW, Australia
| | - Sonu M M Bhaskar
- Global Health Neurology Lab, Sydney, NSW, Australia
- UNSW Medicine and Health, University of New South Wales (UNSW), South West Sydney Clinical Campuses, Sydney, NSW, Australia
- NSW Brain Clot Bank, NSW Health Pathology, Sydney, NSW, Australia
- Department of Neurology & Neurophysiology, Liverpool Hospital and South West Sydney Local Health District, Liverpool, NSW, Australia
- Clinical Sciences Stream, Ingham Institute for Applied Medical Research, Liverpool, NSW, Australia
- Department of Neurology, Division of Cerebrovascular Medicine and Neurology, National Cerebral and Cardiovascular Center (NCVC), Suita, Osaka, Japan
| |
Collapse
|
7
|
Kumar S, Ghosh S, Shanavas N, Sivaramakrishnan V, Dwari M, Das S. Development of pial collaterals by extension of pre-existing artery tips. Cell Rep 2024; 43:114771. [PMID: 39325624 DOI: 10.1016/j.celrep.2024.114771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/02/2024] [Accepted: 09/01/2024] [Indexed: 09/28/2024] Open
Abstract
Pial collaterals provide protection from ischemic damage and improve the prognosis of stroke patients. The origin or precise sequence of events underlying pial collateral development is unclear and has prevented clinicians from adapting new vascularization and regeneration therapies. We use genetic lineage tracing and intravital imaging of mouse brains at cellular resolution to show that during embryogenesis, pial collateral arteries develop from extension and anastomoses of pre-existing artery tips in a VegfR2-dependent manner. This process of artery tip extension occurs on pre-determined microvascular tracks. Our data demonstrate that an arterial receptor, Cxcr4, earlier shown to drive artery cell migration and coronary collateral development, is dispensable for the formation and maintenance of pial collateral arteries. Our study shows that collateral arteries of the brain are built by a mechanism distinct from that of the heart.
Collapse
Affiliation(s)
- Suraj Kumar
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, KA 560065, India
| | - Swarnadip Ghosh
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, KA 560065, India
| | - Niloufer Shanavas
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, KA 560065, India
| | - Vinayak Sivaramakrishnan
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, KA 560065, India
| | - Manish Dwari
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, KA 560065, India
| | - Soumyashree Das
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, KA 560065, India.
| |
Collapse
|
8
|
Brunelli N, Altamura C, Marcosano M, Rossi SS, Costa CM, Fallacara A, Bach-Pages M, Silvestrini M, Mallio CA, Vernieri F. Cerebral vasomotor reactivity in the acute phase and after 6 months in non-disabling stroke/TIA: A prospective cohort study. J Stroke Cerebrovasc Dis 2024; 33:107841. [PMID: 38945417 DOI: 10.1016/j.jstrokecerebrovasdis.2024.107841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/16/2024] [Accepted: 06/27/2024] [Indexed: 07/02/2024] Open
Abstract
BACKGROUND AND AIM Cerebral Vasomotor Reactivity (VMR) is a property of cerebral hemodynamics that protects from cerebrovascular disease. We aimed to explore the VMR longitudinal changes in patients with acute non-disabling stroke/Transient Ischemic Attack (TIA) to understand its implication in stroke ethiopatogenesis. METHODS VMR by Transcranial Doppler Breath Holding test was performed at 48-72 h from stroke onset (T1) and after 6 months (T2) on MCA of the non-affected hemisphere and PCA of the affected hemisphere. RESULTS We consecutively enrolled 124 patients with a median age of 66.0 (IQR 54.75-74.25) years with a median NIHSS 2 (IQR 1-3). Both MCA (1.38 %/s SD 0.58) and PCA (1.35 %/s SD 0.75) BHI at T1 did not differ among different stroke subtypes (p=0.067 and p=0.350; N=124). MCA and PCA BHI decreased from T1 to T2 (respectively 1.39 %/s SD 0.56 vs 1.18%/s SD 0.44 and 1.30 %/s SD 0.69 vs 1.20 %/s SD 0.51; N=109) regardless of ethiopatogenesis (respectively p<0.0001 and p=0.111). CONCLUSION The VMR is higher in acute phase than at 6 months in patients with non-disabling stroke/TIA, regardless of etiopathogenesis. The higher VMR in acute phase could be sustained by an increased Cerebral Blood Flow due to collateral circulation activation supporting the ischemic zone.
Collapse
Affiliation(s)
- Nicoletta Brunelli
- Department of Medicine and Surgery, Unit of Headache and Neurosonology, Unit of Neurology, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21 00128 Roma, Italy; Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200 00128 Roma, Italy.
| | - Claudia Altamura
- Department of Medicine and Surgery, Unit of Headache and Neurosonology, Unit of Neurology, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21 00128 Roma, Italy; Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200 00128 Roma, Italy.
| | - Marilena Marcosano
- Department of Medicine and Surgery, Unit of Headache and Neurosonology, Unit of Neurology, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21 00128 Roma, Italy; Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200 00128 Roma, Italy.
| | - Sergio Soeren Rossi
- Department of Medicine and Surgery, Unit of Headache and Neurosonology, Unit of Neurology, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21 00128 Roma, Italy; Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200 00128 Roma, Italy.
| | | | | | - Marcel Bach-Pages
- Department of Biology, University of Oxford, Oxford OX1 3RB, United Kingdom
| | | | - Carlo Augusto Mallio
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200 00128 Roma, Italy; Unit of Radiology, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21 00128 Rome, Italy.
| | - Fabrizio Vernieri
- Department of Medicine and Surgery, Unit of Headache and Neurosonology, Unit of Neurology, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21 00128 Roma, Italy; Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200 00128 Roma, Italy.
| |
Collapse
|
9
|
LUO S, YANG F, CHEN Y, ZHAO R, LIU H, GAO F, MA W, GAO W, YU W. Sanhua Tang protects against ischemic stroke by preventing blood-brain barrier injury: a network pharmacology and experiments. J TRADIT CHIN MED 2024; 44:794-803. [PMID: 39066540 PMCID: PMC11337263 DOI: 10.19852/j.cnki.jtcm.20240515.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 07/16/2023] [Indexed: 07/28/2024]
Abstract
OBJECTIVE To assess the effect and mechanism of Sanhua Tang (, SHT) in treating ischemic stroke (IS) through the "Kaitong Xuanfu" theory by using network pharmacology and animal experiments. METHODS The active ingredients and targets of SHT and IS were screened by public databases such as Traditional Chinese Medicine systems pharmacology, GeneCards, and online mendelian inheritance in man. Visual network topographies were constructed using R, Cytoscape 3.6.0, AutoDockTools, a user-sponsored molecular visualization system on an open-source foundation, and other software to analyze the correlation between targets and active ingredients. The middle cerebral artery occlusion (MCAO) model was established by operation. Animals were divided into the Sham group, MCAO group (M group), aloe-emodin (AE) group (MCAO rats treated with aloe-emodin), SHT at low dosage (SL group) (MCAO rats treated with SL), SHT at medium dosage (SM group), and SHT at high dosage (SH group). 2,3,5-triphenyl tetrazolium chloride staining was used to detect the volume of cerebral infarction; Nissl staining was used to observe the morphology of neuronal cells; transmission electron microscopy was used to observe the integrity of the blood-brain barrier (BBB); enzyme-linked immunosorbent assay was used to detect the content of interleukin-6 (IL-6), IL-10, tumor necrosis factor α (TNF-α) in serum. Western blot was used to detect the expression of vascular endothelial growth factor A (VEGFA) protein in the cerebral ischemic penumbra. RESULTS Using network pharmacology and molecular docking validation, four active ingredients (lignan, naringenin, aloe-rhodopsin, and β-sitosterol), seven target proteins (protein kinase b 1, IL-6, TNF, VEGFA, TP53, jun proto-oncogene, and cysteinyl aspartate specific proteinase 3), and inflammatory signaling pathways were identified. Animal experiments showed that the SH and AE groups had fewer neurological deficits, reduced brain infarct volumes, decreased serum inflammatory factor levels, increased expression of VEGFA protein, and less structural damage to neurons and BBB. CONCLUSION The present study found that the therapeutic mechanism of SHT against IS may be related to the inhibition of BBB inflammatory damage, which is also the mechanism of "Kaitong Xuanfu." The high-dose group of SHT was relatively effective in regulating inflammatory factors, improving BBB permeability, and protecting neuronal cells from damage.
Collapse
Affiliation(s)
- Shan LUO
- 1 College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Fan YANG
- 2 Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Yuanchun CHEN
- 2 Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Ruoxi ZHAO
- 2 Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Haiye LIU
- 1 College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Fei GAO
- 3 College of Acupuncture and Massage, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Wencan MA
- 3 College of Acupuncture and Massage, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Weijuan GAO
- 1 College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- 4 Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Diseases, Shijiazhuang 050091, China
| | - Wentao YU
- 3 College of Acupuncture and Massage, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- 4 Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Diseases, Shijiazhuang 050091, China
- 5 Hebei International Joint Research Centre for Acupuncture and Moxibustion of Traditional Chinese Medicine, Shijiazhuang 050091, China
| |
Collapse
|
10
|
Zhang Y, Kang Z, Wang J, Liu S, Liu X, Li Z, Li Y, Wang Y, Fu Z, Li J, Huang Y, Ru Z, Peng Y, Yang Z, Wang Y, Yang X, Luo M. Peptide OM-LV20 promotes arteriogenesis induced by femoral artery ligature via the miR-29b-3p/VEGFA axis. Atherosclerosis 2024; 391:117487. [PMID: 38492245 DOI: 10.1016/j.atherosclerosis.2024.117487] [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: 04/07/2023] [Revised: 02/18/2024] [Accepted: 02/22/2024] [Indexed: 03/18/2024]
Abstract
BACKGROUND AND AIMS Therapeutic arteriogenesis is a promising direction for the treatment of ischemic disease caused by atherosclerosis. However, pharmacological or biological approaches to stimulate functional collateral vessels are not yet available. Identifying new drug targets to promote and explore the underlying mechanisms for therapeutic arteriogenesis is necessary. METHODS Peptide OM-LV20 (20 ng/kg) was administered for 7 consecutive days on rat hindlimb ischemia model, collateral vessel growth was assessed by H&E staining, liquid latex perfusion, and specific immunofluorescence. In vitro, we detected the effect of OM-LV20 on human umbilical vein endothelial cells (HUVEC) proliferation and migration. After transfection, we performed quantitative real-time polymerase chain reaction, in situ-hybridization and dual luciferase reporters to assessed effective miRNAs and target genes. The proteins related to downstream signaling pathways were detected by Western blot. RESULTS OM-LV20 significantly increased visible collateral vessels and endothelial nitric oxide synthase (eNOS), together with enhanced inflammation cytokine and monocytes/macrophage infiltration in collateral vessels. In vitro, we defined a novel microRNA (miR-29b-3p), and its inhibition enhanced proliferation and migration of HUVEC, as well as the expression of vascular endothelial growth factor A (VEGFA). OM-LV20 also promoted migration and proliferation of HUVEC, and VEGFA expression was mediated via inhibition of miR-29b-3p. Furthermore, OM-LV20 influenced the protein levels of VEGFR2 and phosphatidylinositol3-kinase (PI3K)/AKT and eNOS in vitro and invivo. CONCLUSIONS Our data indicated that OM-LV20 enhanced arteriogenesis via the miR-29b-3p/VEGFA/VEGFR2-PI3K/AKT/eNOS axis, and highlighte the application potential of exogenous peptide molecular probes through miRNA, which could promote effective therapeutic arteriogenesis in ischemic conditions.
Collapse
Affiliation(s)
- Yingxuan Zhang
- Department of Anatomy & Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Zijian Kang
- Department of Anatomy & Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Jianjun Wang
- School of Clinical Medicine, Xiangnan University, Chenzhou, 423000, Hunan, China
| | - Sahua Liu
- Department of Vascular Surgery, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 571300, Hainan, China
| | - Xin Liu
- Department of Anatomy & Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Zhiruo Li
- Department of Anatomy & Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Yilin Li
- Department of Anatomy & Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Yinglei Wang
- Department of Anatomy & Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Zhe Fu
- Department of Anatomy & Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Jiayi Li
- Department of Anatomy & Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Yubing Huang
- Department of Anatomy & Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Zeqiong Ru
- Department of Anatomy & Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Ying Peng
- Department of Anatomy & Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Zhiyu Yang
- Department of Anatomy & Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Ying Wang
- Key Laboratory of Chemistry in Ethnic Medicinal Resources & Key Laboratory of Natural Products Synthetic Biology of Ethnic Medicinal Endophytes, State Ethnic Affairs Commission & Ministry of Education, School of Ethnic Medicine, Yunnan Minzu University, Kunming, Yunnan, 650504, China.
| | - Xinwang Yang
- Department of Anatomy & Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500, Yunnan, China.
| | - Mingying Luo
- Department of Anatomy & Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500, Yunnan, China.
| |
Collapse
|
11
|
Faber JE, Zhang H, Xenakis JG, Bell TA, Hock P, Pardo-Manuel de Villena F, Ferris MT, Rzechorzek W. Large differences in collateral blood vessel abundance among individuals arise from multiple genetic variants. J Cereb Blood Flow Metab 2023; 43:1983-2004. [PMID: 37572089 PMCID: PMC10676139 DOI: 10.1177/0271678x231194956] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 08/14/2023]
Abstract
Collateral blood flow varies greatly among humans for reasons that remain unclear, resulting in significant differences in ischemic tissue damage. A similarly large variation has also been found in mice that is caused by genetic background-dependent differences in the extent of collateral formation, termed collaterogenesis-a unique angiogenic process that occurs during development and determines collateral number and diameter in the adult. Previous studies have identified several quantitative trait loci (QTL) linked to this variation. However, understanding has been hampered by the use of closely related inbred strains that do not model the wide genetic variation present in the "outbred" human population. The Collaborative Cross (CC) multiparent mouse genetic reference panel was developed to address this limitation. Herein we measured the number and average diameter of cerebral collaterals in 60 CC strains, their 8 founder strains, 8 F1 crosses of CC strains selected for abundant versus sparse collaterals, and 2 intercross populations created from the latter. Collateral number evidenced 47-fold variation among the 60 CC strains, with 14% having poor, 25% poor-to-intermediate, 47% intermediate-to-good, and 13% good collateral abundance, that was associated with large differences in post-stroke infarct volume. Collateral number in skeletal muscle and intestine of selected high- and low-collateral strains evidenced the same relative abundance as in brain. Genome-wide mapping demonstrated that collateral abundance is a highly polymorphic trait. Subsequent analysis identified: 6 novel QTL circumscribing 28 high-priority candidate genes harboring putative loss-of-function polymorphisms (SNPs) associated with low collateral number; 335 predicted-deleterious SNPs present in their human orthologs; and 32 genes associated with vascular development but lacking protein coding variants. Six additional suggestive QTL (LOD > 4.5) were also identified in CC-wide QTL mapping. This study provides a comprehensive set of candidate genes for future investigations aimed at identifying signaling proteins within the collaterogenesis pathway whose variants potentially underlie genetic-dependent collateral insufficiency in brain and other tissues.
Collapse
Affiliation(s)
- James E Faber
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
- Curriculum in Neuroscience, University of North Carolina, Chapel Hill, NC, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Hua Zhang
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
| | - James G Xenakis
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Timothy A Bell
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Pablo Hock
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Fernando Pardo-Manuel de Villena
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
- Carolina Institute for Developmental Disabilities, University of North Carolina, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Martin T Ferris
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Wojciech Rzechorzek
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
12
|
Nagy D, Hricisák L, Walford GP, Lékai Á, Karácsony G, Várbíró S, Ungvári Z, Benyó Z, Pál É. Disruption of Vitamin D Signaling Impairs Adaptation of Cerebrocortical Microcirculation to Carotid Artery Occlusion in Hyperandrogenic Female Mice. Nutrients 2023; 15:3869. [PMID: 37764653 PMCID: PMC10534509 DOI: 10.3390/nu15183869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
Vitamin D deficiency contributes to the pathogenesis of age-related cerebrovascular diseases, including ischemic stroke. Sex hormonal status may also influence the prevalence of these disorders, indicated by a heightened vulnerability among postmenopausal and hyperandrogenic women. To investigate the potential interaction between sex steroids and disrupted vitamin D signaling in the cerebral microcirculation, we examined the cerebrovascular adaptation to unilateral carotid artery occlusion (CAO) in intact, ovariectomized, and hyperandrogenic female mice with normal or functionally inactive vitamin D receptor (VDR). We also analyzed the morphology of leptomeningeal anastomoses, which play a significant role in the compensation. Ablation of VDR by itself did not impact the cerebrocortical adaptation to CAO despite the reduced number of pial collaterals. While ovariectomy did not undermine compensatory mechanisms following CAO, androgen excess combined with VDR inactivity resulted in prolonged hypoperfusion in the cerebral cortex ipsilateral to the occlusion. These findings suggest that the cerebrovascular consequences of disrupted VDR signaling are less pronounced in females, providing a level of protection even after ovariectomy. Conversely, even short-term androgen excess with lacking VDR signaling may lead to unfavorable outcomes of ischemic stroke, highlighting the complex interplay between sex steroids and vitamin D in terms of cerebrovascular diseases.
Collapse
Affiliation(s)
- Dorina Nagy
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (G.P.W.); (Á.L.); (G.K.); (Z.B.)
- Cerebrovascular and Neurocognitive Disorders Research Group, Eötvös Loránd Research Network, Semmelweis University, 1094 Budapest, Hungary
| | - László Hricisák
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (G.P.W.); (Á.L.); (G.K.); (Z.B.)
- Cerebrovascular and Neurocognitive Disorders Research Group, Eötvös Loránd Research Network, Semmelweis University, 1094 Budapest, Hungary
| | - Guillaume Peter Walford
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (G.P.W.); (Á.L.); (G.K.); (Z.B.)
| | - Ágnes Lékai
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (G.P.W.); (Á.L.); (G.K.); (Z.B.)
| | - Gábor Karácsony
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (G.P.W.); (Á.L.); (G.K.); (Z.B.)
| | - Szabolcs Várbíró
- Department of Obstetrics and Gynecology, Semmelweis University, 1082 Budapest, Hungary;
- Department of Obstetrics and Gynecology, University of Szeged, 6725 Szeged, Hungary
- Workgroup for Science Management, Doctoral School, Semmelweis University, 1085 Budapest, Hungary
| | - Zoltán Ungvári
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, 1089 Budapest, Hungary
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Zoltán Benyó
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (G.P.W.); (Á.L.); (G.K.); (Z.B.)
- Cerebrovascular and Neurocognitive Disorders Research Group, Eötvös Loránd Research Network, Semmelweis University, 1094 Budapest, Hungary
| | - Éva Pál
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (G.P.W.); (Á.L.); (G.K.); (Z.B.)
- Cerebrovascular and Neurocognitive Disorders Research Group, Eötvös Loránd Research Network, Semmelweis University, 1094 Budapest, Hungary
| |
Collapse
|
13
|
Faber JE, Zhang H, Xenakis JG, Bell TA, Hock P, de Villena FPM, Ferris MT, Rzechorzek W. Large differences in collateral blood vessel abundance among individuals arise from multiple genetic variants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.28.542633. [PMID: 37398475 PMCID: PMC10312463 DOI: 10.1101/2023.05.28.542633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Collateral blood flow varies greatly among humans for reasons that remain unclear, resulting in significant differences in ischemic tissue damage. A similarly large variation has also been found in mice that is caused by genetic background-dependent differences in the extent of collateral formation, termed collaterogenesis-a unique angiogenic process that occurs during development and determines collateral number and diameter in the adult. Previous studies have identified several quantitative trait loci (QTL) linked to this variation. However, understanding has been hampered by the use of closely related inbred strains that do not model the wide genetic variation present in the "outbred" human population. The Collaborative Cross (CC) multiparent mouse genetic reference panel was developed to address this limitation. Herein we measured the number and average diameter of cerebral collaterals in 60 CC strains, their 8 founder strains, 8 F1 crosses of CC strains selected for abundant versus sparse collaterals, and 2 intercross populations created from the latter. Collateral number evidenced 47-fold variation among the 60 CC strains, with 14% having poor, 25% poor-to-intermediate, 47% intermediate-to-good, and 13% good collateral abundance, that was associated with large differences in post-stroke infarct volume. Genome-wide mapping demonstrated that collateral abundance is a highly polymorphic trait. Subsequent analysis identified: 6 novel QTL circumscribing 28 high-priority candidate genes harboring putative loss-of-function polymorphisms (SNPs) associated with low collateral number; 335 predicted-deleterious SNPs present in their human orthologs; and 32 genes associated with vascular development but lacking protein coding variants. This study provides a comprehensive set of candidate genes for future investigations aimed at identifying signaling proteins within the collaterogenesis pathway whose variants potentially underlie genetic-dependent collateral insufficiency in brain and other tissues.
Collapse
|
14
|
Zhang S, Yu Y, Xu P, Shen X, Fang C, Wu X, Qu P, Wu T, Wang QM, Luo X, Hong Y. Mechanical digit sensory stimulation: a randomized control trial on neurological and motor recovery in acute stroke. Front Neurosci 2023; 17:1134904. [PMID: 37287803 PMCID: PMC10242038 DOI: 10.3389/fnins.2023.1134904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 05/10/2023] [Indexed: 06/09/2023] Open
Abstract
Background Mechanical digit sensory stimulation (MDSS) is a novel therapy designed to accelerate the recovery of upper limb (including hand) function in patients with hemiplegia following a stroke. The primary goal of this study was to investigate the effect of MDSS on patients with acute ischemic stroke (AIS). Methods Sixty-one inpatients with AIS were randomly divided into conventional rehabilitation group (RG) and stimulation group (SG), and the latter group received MDSS therapy. A healthy group consisting of 30 healthy adults was also included. The interleukin-17A (IL-17A), vascular endothelial growth factor A (VEGF-A), and tumor necrosis factor-alpha (TNF-α) plasma levels were measured in all subjects. The neurological and motor functions of patients were evaluated using the National Institutes of Health Stroke Scale (NIHSS), Mini-Mental State Examination (MMSE), Fugel-Meyer Assessment (FMA), and Modified Barthel Index (MBI). Results After 12 days of intervention, the IL-17A, TNF-α, and NIHSS levels were significantly decreased, while the VEGF-A, MMSE, FMA, and MBI levels were significantly increased in both disease groups. No significant difference was observed between both disease groups after intervention. The levels of IL-17A and TNF-α were positively correlated with NIHSS but negatively correlated with MMSE, FMA, and MBI. The VEGF-A levels were negatively correlated with NIHSS but positively correlated with MMSE, FMA, and MBI. Conclusion Both MDSS and conventional rehabilitation significantly reduce the production of IL-17A and TNF-α, increase the VEGF-A levels, and effectively improve cognition and motor function of hemiplegic patients with AIS, and the effects of MDSS and conventional rehabilitation are comparable.
Collapse
Affiliation(s)
- Shuting Zhang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Yang Yu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Panpan Xu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Xianshan Shen
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Chuanqin Fang
- Department of Neurology, The Second Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Xiaosan Wu
- Department of Neurology, The Second Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Ping Qu
- Department of Neurology, The Second Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Tingting Wu
- Key Laboratory of Oral Disease Research of Anhui Province, Stomatologic Hospital and College, Anhui Medical University, Hefei, Anhui Province, China
| | - Qing Mei Wang
- Stroke Biological Recovery Laboratory, Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA, United States
| | - Xun Luo
- School of Medicine, Shenzhen University, Shenzhen, Guangdong Province, China
| | - Yongfeng Hong
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| |
Collapse
|
15
|
Chen Q, Li Y, Bie B, Zhao B, Zhang Y, Fang S, Li S, Zhang Y. P38 MAPK activated ADAM17 mediates ACE2 shedding and promotes cardiac remodeling and heart failure after myocardial infarction. Cell Commun Signal 2023; 21:73. [PMID: 37046278 PMCID: PMC10091339 DOI: 10.1186/s12964-023-01087-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 02/23/2023] [Indexed: 04/14/2023] Open
Abstract
BACKGROUND Heart failure (HF) after myocardial infarction (MI) is a prevalent disease with a poor prognosis. Relieving pathological cardiac remodeling and preserving cardiac function is a critical link in the treatment of post-MI HF. Thus, more new therapeutic targets are urgently needed. The expression of ADAM17 is increased in patients with acute MI, but its functional role in post-MI HF remains unclear. METHODS To address this question, we examined the effects of ADAM17 on the severity and prognosis of HF within 1 year of MI in 152 MI patients with or without HF. In mechanistic studies, the effects of ADAM17 on ventricular remodeling and systolic function were extensively assessed at the tissue and cellular levels by establishing animal model of post-MI HF and in vitro hypoxic cell model. RESULTS High levels of ADAM17 predicted a higher incidence of post-MI HF, poorer cardiac function and higher mortality. Animal studies demonstrated that ADAM17 promoted the occurrence of post-MI HF, as indicated by increased infarct size, cardiomyocyte hypertrophy, myocardial interstitial collagen deposition and cardiac failure. ADAM17 knock down significantly improved pathological cardiac remodeling and cardiac function in mice with MI. Mechanistically, activated ADAM17 inhibited the cardioprotective effects of ACE2 by promoting hydrolytic shedding of the transmembrane protein ACE2 in cardiomyocytes, which subsequently mediated the occurrence of cardiac remodeling and the progression of heart failure. Moreover, the activation of ADAM17 in hypoxic cardiomyocytes was dependent on p38 MAPK phosphorylation at threonine 735. CONCLUSIONS These data highlight a novel and important mechanism for ADAM17 to cause post-MI HF, which will hopefully be a new potential target for early prediction or intervention of post-MI HF. Video abstract.
Collapse
Affiliation(s)
- Qi Chen
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Road, Nangang District, Harbin, 150001, Heilongjiang Province, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, No. 246 Xuefu Road, Nangang District, Harbin, 150001, Heilongjiang Province, China
- Harbin Medical University, No. 157 JianBao Road, Nangang District, Harbin, 150001, Heilongjiang Province, China
| | - Yilan Li
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Road, Nangang District, Harbin, 150001, Heilongjiang Province, China
| | - Bike Bie
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Road, Nangang District, Harbin, 150001, Heilongjiang Province, China
- Harbin Medical University, No. 157 JianBao Road, Nangang District, Harbin, 150001, Heilongjiang Province, China
| | - Bin Zhao
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Road, Nangang District, Harbin, 150001, Heilongjiang Province, China
- Harbin Medical University, No. 157 JianBao Road, Nangang District, Harbin, 150001, Heilongjiang Province, China
| | - Yanxiu Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Road, Nangang District, Harbin, 150001, Heilongjiang Province, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, No. 246 Xuefu Road, Nangang District, Harbin, 150001, Heilongjiang Province, China
- Harbin Medical University, No. 157 JianBao Road, Nangang District, Harbin, 150001, Heilongjiang Province, China
| | - Shaohong Fang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Road, Nangang District, Harbin, 150001, Heilongjiang Province, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, No. 246 Xuefu Road, Nangang District, Harbin, 150001, Heilongjiang Province, China
| | - Shuijie Li
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Yao Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Road, Nangang District, Harbin, 150001, Heilongjiang Province, China.
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, No. 246 Xuefu Road, Nangang District, Harbin, 150001, Heilongjiang Province, China.
| |
Collapse
|
16
|
Tian Y, Fopiano KA, Buncha V, Lang L, Suggs HA, Wang R, Rudic RD, Filosa JA, Bagi Z. The role of ADAM17 in cerebrovascular and cognitive function in the APP/PS1 mouse model of Alzheimer's disease. Front Mol Neurosci 2023; 16:1125932. [PMID: 36937050 PMCID: PMC10018024 DOI: 10.3389/fnmol.2023.1125932] [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: 12/16/2022] [Accepted: 01/27/2023] [Indexed: 03/06/2023] Open
Abstract
Introduction The disintegrin and metalloproteinase 17 (ADAM17) exhibits α-secretase activity, whereby it can prevent the production of neurotoxic amyloid precursor protein-α (APP). ADAM17 is abundantly expressed in vascular endothelial cells and may act to regulate vascular homeostatic responses, including vasomotor function, vascular wall morphology, and formation of new blood vessels. The role of vascular ADAM17 in neurodegenerative diseases remains poorly understood. Here, we hypothesized that cerebrovascular ADAM17 plays a role in the pathogenesis of Alzheimer's disease (AD). Methods and results We found that 9-10 months old APP/PS1 mice with b-amyloid accumulation and short-term memory and cognitive deficits display a markedly reduced expression of ADAM17 in cerebral microvessels. Systemic delivery and adeno-associated virus (AAV)-mediated re-expression of ADAM17 in APP/PS1 mice improved cognitive functioning, without affecting b-amyloid plaque density. In isolated and pressurized cerebral arteries of APP/PS1 mice the endothelium-dependent dilation to acetylcholine was significantly reduced, whereas the vascular smooth muscle-dependent dilation to the nitric oxide donor, sodium nitroprusside was maintained when compared to WT mice. The impaired endothelium-dependent vasodilation of cerebral arteries in APP/PS1 mice was restored to normal level by ADAM17 re-expression. The cerebral artery biomechanical properties (wall stress and elasticity) and microvascular network density was not affected by ADAM17 re-expression in the APP/PS1 mice. Additionally, proteomic analysis identified several differentially expressed molecules involved in AD neurodegeneration and neuronal repair mechanisms that were reversed by ADAM17 re-expression. Discussion Thus, we propose that a reduced ADAM17 expression in cerebral microvessels impairs vasodilator function, which may contribute to the development of cognitive dysfunction in APP/PS1 mice, and that ADAM17 can potentially be targeted for therapeutic intervention in AD.
Collapse
Affiliation(s)
- Yanna Tian
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Katie Anne Fopiano
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Vadym Buncha
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Liwei Lang
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Hayden A. Suggs
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Rongrong Wang
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - R. Daniel Rudic
- Department of Pharmacology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Jessica A. Filosa
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Zsolt Bagi
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
17
|
Tian Y, Niu HT, Li MH, Wang YZ. Effect of VEGF on neurological impairment and prognosis of acute cerebral infarction patients: A retrospective case-control study. Medicine (Baltimore) 2023; 102:e29835. [PMID: 36820574 PMCID: PMC9907990 DOI: 10.1097/md.0000000000029835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2023] Open
Abstract
OBJECTIVE Due to the complex pathological mechanism of acute cerebral infarction, the role of vascular endothelial growth factor (VEGF) on the disease is not clear. Therefore, a retrospective case-control study was performed to explore the effect of VEGF on neurological impairment and prognosis of acute cerebral infarction patients. METHOD A total of 100 patients with acute cerebral infarction admitted to our hospital from April 2021 to April 2022 were selected. Blood samples from all patients would be routinely collected to detect the expression of serum VEGF. Pearson chi-square, Spearman correlation and univariate Logistic regression were used to analyze the clinical data to explore the relationship between VEGF expression and basic information, stroke degree, quality of life, and prognosis of patients. To determine whether VEGF can provide relevant basis for the early prevention and prognostic treatment of acute cerebral infarction. And multivariate logistic regression was used to calculate the odds ratio between each variable and VEGF expression. RESULTS Pearson chi-square test and Spearman correlation coefficient showed that sex, degree of stroke, limb convulsions, loss of consciousness, hemiplegia, aphasia, mental functioning score, overall quality of life score, and short-term prognosis were significantly correlated with VEGF expression in 100 patients. Univariate logistic regression was used to describe the ORs and 95% confidence interval of subjects at the univariate level, and the degree of stroke (OR = 83.333, P < 0.001), tic of limbs (OR = 26.316, P < 0.001), loss of consciousness (OR = 23.256, P < 0.001), hemiplegia (OR = 62.500, P < 0.001), aphasia (OR = 76.923, P < 0.001), mental functioning score (OR = 7.937, P < 0.001), overall quality of life score (OR = 5.464, P < 0.001), short-term prognosis (OR = 37.037, P < 0.001) was significantly correlated with the high expression of VEGF. CONCLUSIONS The level of serum VEGF was positively correlated with neurological impairment degree and prognosis in patients with acute cerebral infarction, the more severe the degree of stroke and the worse the prognosis.
Collapse
Affiliation(s)
- Yong Tian
- Department of neurosurgery, Cangzhou Central Hospital, Cangzhou, Hebei province, P.R. China
- * Correspondence: Yong Tian, Department of neurosurgery, Cangzhou Central Hospital, No. 16 Xinhua Western Road, Cangzhou, Hebei 061000, P.R. China (e-mail: )
| | - Hai-Tao Niu
- Department of neurosurgery, Cangzhou Central Hospital, Cangzhou, Hebei province, P.R. China
| | - Ming-Hang Li
- Department of neurosurgery, Cangzhou Central Hospital, Cangzhou, Hebei province, P.R. China
| | - Yang-Zhou Wang
- Department of neurosurgery, Cangzhou Central Hospital, Cangzhou, Hebei province, P.R. China
| |
Collapse
|
18
|
Kim CK, Hwang JY, Hong TH, Lee DM, Lee K, Nam H, Joo KM. Combination stem cell therapy using dental pulp stem cells and human umbilical vein endothelial cells for critical hindlimb ischemia. BMB Rep 2022. [PMID: 35168701 PMCID: PMC9340082 DOI: 10.5483/bmbrep.2022.55.7.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Narrowing of arteries supplying blood to the limbs provokes critical hindlimb ischemia (CLI). Although CLI results in irreversible sequelae, such as amputation, few therapeutic options induce the formation of new functional blood vessels. Based on the proangiogenic potentials of stem cells, in this study, it was examined whether a combination of dental pulp stem cells (DPSCs) and human umbilical vein endothelial cells (HUVECs) could result in enhanced therapeutic effects of stem cells for CLI compared with those of DPSCs or HUVECs alone. The DPSCs+ HUVECs combination therapy resulted in significantly higher blood flow and lower ischemia damage than DPSCs or HUVECs alone. The improved therapeutic effects in the DPSCs+ HUVECs group were accompanied by a significantly higher number of microvessels in the ischemic tissue than in the other groups. In vitro proliferation and tube formation assay showed that VEGF in the conditioned media of DPSCs induced proliferation and vessel-like tube formation of HUVECs. Altogether, our results demonstrated that the combination of DPSCs and HUVECs had significantly better therapeutic effects on CLI via VEGF-mediated crosstalk. This combinational strategy could be used to develop novel clinical protocols for CLI proangiogenic regenerative treatments.
Collapse
Affiliation(s)
- Chung Kwon Kim
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea
- Medical Innovation Technology Inc. (MEDINNO Inc.), Seoul 08517, Korea
| | - Ji-Yoon Hwang
- Medical Innovation Technology Inc. (MEDINNO Inc.), Seoul 08517, Korea
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16149, Korea
| | - Tae Hee Hong
- Medical Innovation Technology Inc. (MEDINNO Inc.), Seoul 08517, Korea
| | - Du Man Lee
- Medical Innovation Technology Inc. (MEDINNO Inc.), Seoul 08517, Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Korea
| | - Kyunghoon Lee
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16149, Korea
| | - Hyun Nam
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16149, Korea
- Stem Cell and Regenerative Medicine Institute, Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Korea
- Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon 16149, Korea
| | - Kyeung Min Joo
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea
- Medical Innovation Technology Inc. (MEDINNO Inc.), Seoul 08517, Korea
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16149, Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Korea
- Stem Cell and Regenerative Medicine Institute, Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Korea
| |
Collapse
|
19
|
Kim CK, Hwang JY, Hong TH, Lee DM, Lee K, Nam H, Joo KM. Combination stem cell therapy using dental pulp stem cells and human umbilical vein endothelial cells for critical hindlimb ischemia. BMB Rep 2022; 55:336-341. [PMID: 35168701 PMCID: PMC9340082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 01/25/2022] [Accepted: 02/11/2022] [Indexed: 03/08/2024] Open
Abstract
Narrowing of arteries supplying blood to the limbs provokes critical hindlimb ischemia (CLI). Although CLI results in irreversible sequelae, such as amputation, few therapeutic options induce the formation of new functional blood vessels. Based on the proangiogenic potentials of stem cells, in this study, it was examined whether a combination of dental pulp stem cells (DPSCs) and human umbilical vein endothelial cells (HUVECs) could result in enhanced therapeutic effects of stem cells for CLI compared with those of DPSCs or HUVECs alone. The DPSCs+ HUVECs combination therapy resulted in significantly higher blood flow and lower ischemia damage than DPSCs or HUVECs alone. The improved therapeutic effects in the DPSCs+ HUVECs group were accompanied by a significantly higher number of microvessels in the ischemic tissue than in the other groups. In vitro proliferation and tube formation assay showed that VEGF in the conditioned media of DPSCs induced proliferation and vessel-like tube formation of HUVECs. Altogether, our results demonstrated that the combination of DPSCs and HUVECs had significantly better therapeutic effects on CLI via VEGF-mediated crosstalk. This combinational strategy could be used to develop novel clinical protocols for CLI proangiogenic regenerative treatments. [BMB Reports 2022; 55(7): 336-341].
Collapse
Affiliation(s)
- Chung Kwon Kim
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea
- Medical Innovation Technology Inc. (MEDINNO Inc.), Seoul 08517, Korea
| | - Ji-Yoon Hwang
- Medical Innovation Technology Inc. (MEDINNO Inc.), Seoul 08517, Korea
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16149, Korea
| | - Tae Hee Hong
- Medical Innovation Technology Inc. (MEDINNO Inc.), Seoul 08517, Korea
| | - Du Man Lee
- Medical Innovation Technology Inc. (MEDINNO Inc.), Seoul 08517, Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Korea
| | - Kyunghoon Lee
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16149, Korea
| | - Hyun Nam
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16149, Korea
- Stem Cell and Regenerative Medicine Institute, Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Korea
- Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon 16149, Korea
| | - Kyeung Min Joo
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea
- Medical Innovation Technology Inc. (MEDINNO Inc.), Seoul 08517, Korea
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16149, Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Korea
- Stem Cell and Regenerative Medicine Institute, Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Korea
- Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon 16149, Korea
| |
Collapse
|
20
|
le Noble F, Kupatt C. Interdependence of Angiogenesis and Arteriogenesis in Development and Disease. Int J Mol Sci 2022; 23:ijms23073879. [PMID: 35409246 PMCID: PMC8999596 DOI: 10.3390/ijms23073879] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/22/2022] [Accepted: 03/27/2022] [Indexed: 02/04/2023] Open
Abstract
The structure of arterial networks is optimized to allow efficient flow delivery to metabolically active tissues. Optimization of flow delivery is a continuous process involving synchronization of the structure and function of the microcirculation with the upstream arterial network. Risk factors for ischemic cardiovascular diseases, such as diabetes mellitus and hyperlipidemia, adversely affect endothelial function, induce capillary regression, and disrupt the micro- to macrocirculation cross-talk. We provide evidence showing that this loss of synchronization reduces arterial collateral network recruitment upon arterial stenosis, and the long-term clinical outcome of current revascularization strategies in these patient cohorts. We describe mechanisms and signals contributing to synchronized growth of micro- and macrocirculation in development and upon ischemic challenges in the adult organism and identify potential therapeutic targets. We conclude that a long-term successful revascularization strategy should aim at both removing obstructions in the proximal part of the arterial tree and restoring “bottom-up” vascular communication.
Collapse
Affiliation(s)
- Ferdinand le Noble
- Department of Cell and Developmental Biology, Institute of Zoology (ZOO), Karlsruhe Institute of Technology (KIT), Fritz Haber Weg 4, 76131 Karlsruhe, Germany
- Institute for Biological and Chemical Systems—Biological Information Processing, Karlsruhe Institute of Technology (KIT), P.O. Box 3640, 76021 Karlsruhe, Germany
- Institute of Experimental Cardiology, Heidelberg Germany and German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, University of Heidelberg, 69117 Heidelberg, Germany
- Correspondence: (F.l.N.); (C.K.)
| | - Christian Kupatt
- Klinik und Poliklinik für Innere Medizin I, Klinikum Rechts der Isar, Technical University Munich, 81675 Munich, Germany
- DZHK (German Center for Cardiovascular Research), Munich Heart Alliance, 80802 Munich, Germany
- Correspondence: (F.l.N.); (C.K.)
| |
Collapse
|
21
|
Perovic T, Harms C, Gerhardt H. Formation and Maintenance of the Natural Bypass Vessels of the Brain. Front Cardiovasc Med 2022; 9:778773. [PMID: 35391845 PMCID: PMC8980479 DOI: 10.3389/fcvm.2022.778773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 02/28/2022] [Indexed: 11/18/2022] Open
Abstract
Ischemic diseases are the leading cause of death and disability worldwide. The main compensatory mechanism by which our body responds to reduced or blocked blood flow caused by ischemia is mediated by collateral vessels. Collaterals are present in many healthy tissues (including brain and heart) and serve as natural bypass vessels, by bridging adjacent arterial trees. This review focuses on: the definition and significance of pial collateral vessels, the described mechanism of pial collateral formation, an overview of molecular players and pathways involved in pial collateral biology and emerging approaches to prevent or mitigate risk factor-associated loss of pial collaterals. Despite their high clinical relevance and recent scientific efforts toward understanding collaterals, much of the fundamental biology of collaterals remains obscure.
Collapse
Affiliation(s)
- Tijana Perovic
- Integrative Vascular Biology, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- *Correspondence: Tijana Perovic
| | - Christoph Harms
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Center for Stroke Research Berlin with Department of Experimental Neurology, Charité Universitaetsmedizin Berlin, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Charité-Universitaetsmedizin Berlin, Berlin, Germany
| | - Holger Gerhardt
- Integrative Vascular Biology, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Holger Gerhardt
| |
Collapse
|
22
|
Ng MA. Posterior Circulation Ischaemic Stroke. Am J Med Sci 2022; 363:388-398. [PMID: 35104439 DOI: 10.1016/j.amjms.2021.10.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 06/09/2021] [Accepted: 10/21/2021] [Indexed: 11/16/2022]
Abstract
Posterior circulation ischaemic stroke (PCIS) is a disease of high burden. They account for 20-25% of all ischaemic strokes. However, it is relatively under-researched and requires more clinical attention, since it carries worse functional outcomes. Vertigo, visual disturbances and sensory/motor disturbances are found in PCIS. Large artery atherosclerosis and embolism are main causes of PCIS, while there is growing evidence that vertebrobasilar dolichoectasia is a key association. Hypertension is the commonest risk factor, while diabetes mellitus is more specific to PCIS. PCIS is diagnosed through neuroimaging techniques, which examine structural brain abnormalities, vascular patency and perfusion. PCIS, in line with ischaemic stroke in general, requires medical treatment and lifestyle modifications. This includes smoking cessation, weight control, and dietary alterations. Aspirin use also significantly improves survival outcomes. While intravascular and intra-arterial thrombolysis improve clinical outcomes, this is not proven conclusively for stenting and angioplasty. Future research on PCIS can focus on multi-centre epidemiological studies, clinically significant anatomical variants, and collateralisation.
Collapse
Affiliation(s)
- Mr Alexander Ng
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Full Address: Block K, Queen Mary Hospital, Pokfulam Road, Hong Kong.
| |
Collapse
|
23
|
ADAM and ADAMTS disintegrin and metalloproteinases as major factors and molecular targets in vascular malfunction and disease. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 94:255-363. [PMID: 35659374 PMCID: PMC9231755 DOI: 10.1016/bs.apha.2021.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
A Disintegrin and Metalloproteinase (ADAM) and A Disintegrin and Metalloproteinase with Thrombospondin Motifs (ADAMTS) are two closely related families of proteolytic enzymes. ADAMs are largely membrane-bound enzymes that act as molecular scissors or sheddases of membrane-bound proteins, growth factors, cytokines, receptors and ligands, whereas ADAMTS are mainly secreted enzymes. ADAMs have a pro-domain, and a metalloproteinase, disintegrin, cysteine-rich and transmembrane domain. Similarly, ADAMTS family members have a pro-domain, and a metalloproteinase, disintegrin, and cysteine-rich domain, but instead of a transmembrane domain they have thrombospondin motifs. Most ADAMs and ADAMTS are activated by pro-protein convertases, and can be regulated by G-protein coupled receptor agonists, Ca2+ ionophores and protein kinase C. Activated ADAMs and ADAMTS participate in numerous vascular processes including angiogenesis, vascular smooth muscle cell proliferation and migration, vascular cell apoptosis, cell survival, tissue repair, and wound healing. ADAMs and ADAMTS also play a role in vascular malfunction and cardiovascular diseases such as hypertension, atherosclerosis, coronary artery disease, myocardial infarction, heart failure, peripheral artery disease, and vascular aneurysm. Decreased ADAMTS13 is involved in thrombotic thrombocytopenic purpura and microangiopathies. The activity of ADAMs and ADAMTS can be regulated by endogenous tissue inhibitors of metalloproteinases and other synthetic small molecule inhibitors. ADAMs and ADAMTS can be used as diagnostic biomarkers and molecular targets in cardiovascular disease, and modulators of ADAMs and ADAMTS activity may provide potential new approaches for the management of cardiovascular disorders.
Collapse
|
24
|
Kulkarni R, Andraska E, McEnaney R. Structural Remodeling of the Extracellular Matrix in Arteriogenesis: A Review. Front Cardiovasc Med 2021; 8:761007. [PMID: 34805316 PMCID: PMC8602576 DOI: 10.3389/fcvm.2021.761007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 10/11/2021] [Indexed: 01/10/2023] Open
Abstract
Lower extremity arterial occlusive disease (AOD) results in significant morbidity and mortality for the population, with up to 10% of patients ultimately requiring amputation. An alternative method for non-surgical revascularization which is yet to be fully understood is the optimization of the body's own natural collateral arterial network in a process known as arteriogenesis. Under conditions of conductance vessel stenosis or occlusion resulting in increased flow, shear forces, and pressure gradients within collaterals, positive remodeling occurs to increase the diameter and capacity of these vessels. The creation of a distal arteriovenous fistula (AVF) will drive increased arteriogenesis as compared to collateral formation with the occlusion of a conductance vessel alone by further increasing flow through these arterioles, demonstrating the capacity for arteriogenesis to form larger, more efficient collaterals beyond what is spontaneously achieved after arterial occlusion. Arteries rely on an extracellular matrix (ECM) composed of elastic fibers and collagens that provide stability under hemodynamic stress, and ECM remodeling is necessary to allow for increased diameter and flow conductance in mature arterial structures. When positive remodeling occurs, digestion of lamella and the internal elastic lamina (IEL) by matrix metalloproteinases (MMPs) and other elastases results in the rearrangement and thinning of elastic structures and may be replaced with disordered elastin synthesis without recovery of elastic function. This results in transmission of wall strain to collagen and potential for aneurysmal degeneration along collateral networks, as is seen in the pancreaticoduodenal artery (PDA) after celiac occlusion and inferior mesenteric artery (IMA) with concurrent celiac and superior mesenteric artery (SMA) occlusions. Further understanding into the development of collaterals is required to both better understand aneurysmal degeneration and optimize collateral formation in AOD.
Collapse
Affiliation(s)
- Rohan Kulkarni
- Division of Vascular Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Elizabeth Andraska
- Division of Vascular Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Ryan McEnaney
- Division of Vascular Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Veterans Affairs Hospitals Pittsburgh Healthcare System, Pittsburgh, PA, United States
| |
Collapse
|
25
|
Liu Y, Long L, Zhang F, Hu X, Zhang J, Hu C, Wang Y, Xu J. Microneedle-mediated vascular endothelial growth factor delivery promotes angiogenesis and functional recovery after stroke. J Control Release 2021; 338:610-622. [PMID: 34481025 DOI: 10.1016/j.jconrel.2021.08.057] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 08/02/2021] [Accepted: 08/30/2021] [Indexed: 02/05/2023]
Abstract
Ischemic stroke is still the major cause of disability worldwide. Although vascular endothelial growth factor (VEGF) is able to promote both angiogenesis and functional recovery, its use is limited by needle-induced injury, nonhomogenous VEGF distribution, and limited VEGF retention in the brain after intracranial or intravenous injection. Here, we first present a gelatin methacryloyl (GelMA) microneedle (MN)-based platform for the sustained and controlled local delivery of an adeno-associated virus (AAV) expressing human VEGF (AAV-VEGF) that achieves homogenous distribution and high transfection efficiency in ischemic brains. An ischemic stroke model was established in adult rats, and MNs loaded with AAV-VEGF were epicortically inserted into both the ischemic core and penumbra of these rats one day after the onset of ischemia. One week later, the inflammatory response and microneedle biocompatibility were assessed by enzyme-linked immunosorbent assay (ELISA) and immunofluorescence. Eight weeks later, angiogenesis and neural stem cell proliferation and migration were assessed. GelMA MN implantation did not elicit an obvious inflammatory response and had good biocompatibility in the brain. AAV-green fluorescent protein (GFP)-loaded MNs could achieve successful transfection and homogeneous distribution in the brain cortex three weeks postoperatively. MNs loaded with AAV-VEGF increased VEGF expression and enhanced functional angiogenesis and neurogenesis. In summary, MNs might emerge as a promising platform for delivering various therapeutics to treat ischemic stroke and repair other neurologically diseased tissues.
Collapse
Affiliation(s)
- Yang Liu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Linyu Long
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Fanjun Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Xuefeng Hu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Jieyu Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Cheng Hu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China.
| | - Jianguo Xu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
26
|
Li Z, Wang M, Gu J, Zhao L, Guo Y, Zhang Z, Liu X. Missense Variants in Hypoxia-Induced VEGFA/VEGFR2 Signaling Predict the Outcome of Large Artery Atherosclerotic Stroke. Cell Mol Neurobiol 2021; 41:1217-1225. [PMID: 32506171 PMCID: PMC11448691 DOI: 10.1007/s10571-020-00890-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 05/28/2020] [Indexed: 11/30/2022]
Abstract
Collateral density variations are a major determinant of stroke outcome. Here, we explored the association of missense variants in hypoxia-induced VEGFA/VEGFR2 signaling and stroke outcome. We recruited 683 large artery atherosclerotic (LAA) stroke patients as the training set from Nanjing Stroke Registry Program between August 2013 and January 2016. To validate the findings from the training set, we recruited an additional 333 LAA stroke patients between February 2016 and January 2017 as the validation set. Genotyping of target SNPs (rs11549465 [HIF-1α], rs11549467 [HIF-1α], rs1870377 [VEGFR2], and rs2305948 [VEGFR2]) was conducted using a SNPscan method. Unfavorable outcome was defined as a modified Rankin Scale (mRS) score > 2 at three months after index event. In the training set, the AA genotype of rs1870377 led to a decreased risk of unfavorable outcomes in the recessive model (AA vs. TA + TT, OR 0.60, 95% CI 0.38-0.95, P = 0.031). This was confirmed in the validation set (OR 0.43, 95% CI 0.21-0.86, P = 0.017) and the combined set (OR 0.54, 95% CI 0.36-0.79, P = 0.002). We also found that A allele was a protective factor for stroke outcome in both validation set and combined set (OR 0.70, 95% CI 0.49-0.99, P = 0.044 and OR 0.77, 95% CI 0.63-0.94, P = 0.012, respectively). In silico analysis indicated that the rs1870377 variant led to structural alterations in VEGFR2 that may influence its activity. Our findings demonstrate that the rs1870377 in the hypoxia-induced VEGFA/VEGFR2 axis predicts the 3-month outcome of patients with LAA stroke.
Collapse
Affiliation(s)
- Zibao Li
- Department of Neurology, Jinling Hospital, Nanjing Medical University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China
- Department of Neurology, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, China
| | - Mengmeng Wang
- Department of Neurology, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, China
| | - Jinyu Gu
- Department of Neurology, Jinling Hospital, Nanjing Medical University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Li Zhao
- Department of Neurology, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, China
| | - Yongtao Guo
- Department of Neurology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, 223300, China
| | - Zhizhong Zhang
- Department of Neurology, Jinling Hospital, Nanjing Medical University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China.
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China.
| | - Xinfeng Liu
- Department of Neurology, Jinling Hospital, Nanjing Medical University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China.
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China.
| |
Collapse
|
27
|
Kawai T, Elliott KJ, Scalia R, Eguchi S. Contribution of ADAM17 and related ADAMs in cardiovascular diseases. Cell Mol Life Sci 2021; 78:4161-4187. [PMID: 33575814 PMCID: PMC9301870 DOI: 10.1007/s00018-021-03779-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/23/2020] [Accepted: 01/27/2021] [Indexed: 02/06/2023]
Abstract
A disintegrin and metalloproteases (ADAMs) are key mediators of cell signaling by ectodomain shedding of various growth factors, cytokines, receptors and adhesion molecules at the cellular membrane. ADAMs regulate cell proliferation, cell growth, inflammation, and other regular cellular processes. ADAM17, the most extensively studied ADAM family member, is also known as tumor necrosis factor (TNF)-α converting enzyme (TACE). ADAMs-mediated shedding of cytokines such as TNF-α orchestrates immune system or inflammatory cascades and ADAMs-mediated shedding of growth factors causes cell growth or proliferation by transactivation of the growth factor receptors including epidermal growth factor receptor. Therefore, increased ADAMs-mediated shedding can induce inflammation, tissue remodeling and dysfunction associated with various cardiovascular diseases such as hypertension and atherosclerosis, and ADAMs can be a potential therapeutic target in these diseases. In this review, we focus on the role of ADAMs in cardiovascular pathophysiology and cardiovascular diseases. The main aim of this review is to stimulate new interest in this area by highlighting remarkable evidence.
Collapse
Affiliation(s)
- Tatsuo Kawai
- Cardiovascular Research Center, Lewis Katz School of Medicine At Temple University, Philadelphia, PA, USA
| | - Katherine J Elliott
- Cardiovascular Research Center, Lewis Katz School of Medicine At Temple University, Philadelphia, PA, USA
| | - Rosario Scalia
- Cardiovascular Research Center, Lewis Katz School of Medicine At Temple University, Philadelphia, PA, USA
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine At Temple University, Philadelphia, PA, USA.
| |
Collapse
|
28
|
Faber JE, Storz JF, Cheviron ZA, Zhang H. High-altitude rodents have abundant collaterals that protect against tissue injury after cerebral, coronary and peripheral artery occlusion. J Cereb Blood Flow Metab 2021; 41:731-744. [PMID: 32703056 PMCID: PMC7983333 DOI: 10.1177/0271678x20942609] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/03/2020] [Accepted: 06/22/2020] [Indexed: 12/22/2022]
Abstract
Collateral number/density varies widely in brain and other tissues among strains of Mus musculus mice due to differences in genetic background. Recent studies have shown that prolonged exposure to reduced atmospheric oxygen induces additional collaterals to form, suggesting that natural selection may favor increased collaterals in populations native to high-altitude. High-altitude guinea pigs (Cavia) and deer mice (Peromyscus) were compared with lowland species of Peromyscus, Mus and Rattus (9 species/strains examined). Collateral density, diameter and other morphometrics were measured in brain where, importantly, collateral abundance reflects that in other tissues of the same individual. Guinea pigs and high-altitude deer mice had a greater density of pial collaterals than lowlanders. Consistent with this, guinea pigs and highlander mice evidenced complete and 80% protection against stroke, respectively. They also sustained significantly less ischemia in heart and lower extremities after arterial occlusion. Vessels of the circle of Willis, including the communicating collateral arteries, also exhibited unique features in the highland species. Our findings support the hypothesis that species native to high-altitude have undergone genetic selection for abundant collaterals, suggesting that besides providing protection in obstructive disease, collaterals serve a physiological function to optimize oxygen delivery to meet oxygen demand when oxygen is limiting.
Collapse
Affiliation(s)
- James E Faber
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jay F Storz
- School of Biological Sciences, University of Nebraska, Lincoln, NE, USA
| | - Zachary A Cheviron
- Division of Biological Sciences, University of Montana, Missoula, MT, USA
| | - Hua Zhang
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
29
|
Aghajanian A, Zhang H, Buckley BK, Wittchen ES, Ma WY, Faber JE. Decreased inspired oxygen stimulates de novo formation of coronary collaterals in adult heart. J Mol Cell Cardiol 2021; 150:1-11. [PMID: 33038388 PMCID: PMC7855913 DOI: 10.1016/j.yjmcc.2020.09.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/13/2020] [Accepted: 09/25/2020] [Indexed: 01/18/2023]
Abstract
RATIONALE Collateral vessels lessen myocardial ischemia when acute or chronic coronary obstruction occurs. It has long been assumed that although native (pre-existing) collaterals enlarge in obstructive disease, new collaterals do not form in the adult. However, the latter was recently shown to occur after coronary artery ligation. Understanding the signals that drive this process is challenged by the difficulty in studying collateral vessels directly and the complex milieu of signaling pathways, including cell death, induced by ligation. Herein we show that hypoxemia alone is capable of inducing collateral vessels to form and that the novel gene Rabep2 is required. OBJECTIVE Hypoxia stimulates angiogenesis during embryonic development and in pathological states. We hypothesized that hypoxia also stimulates collateral formation in adult heart by a process that involves RABEP2, a recently identified protein required for formation of collateral vessels during development. METHODS AND RESULTS Exposure of mice to reduced FiO2 induced collateral formation that resulted in smaller infarctions following LAD ligation and that reversed on return to normoxia. Deletion of Rabep2 or knockdown of Vegfa inhibited formation. Hypoxia upregulated Rabep2, Vegfa and Vegfr2 in heart and brain microvascular endothelial cells (HBMVECs). Knockdown of Rabep2 impaired migration of HBMVECs. In contrast to systemic hypoxia, deletion of Rabep2 did not affect collateral formation induced by ischemic injury caused by LAD ligation. CONCLUSIONS Hypoxia induced formation of coronary collaterals by a process that required VEGFA and RABEP2, proteins also required for collateral formation during development. Knockdown of Rabep2 impaired cell migration, providing one potential mechanism for RABEP2's role in collateral formation. This appears specific to hypoxia, since formation after acute ischemic injury was unaffected in Rabep2-/- mice. These findings provide a novel model for studying coronary collateral formation, and demonstrate that hypoxia alone can induce new collaterals to form in adult heart.
Collapse
Affiliation(s)
- Amir Aghajanian
- Department of Cell Biology and Physiology and the McAllister Heart Institute, University of North Carolina at Chapel Hill, United States of America
| | - Hua Zhang
- Department of Cell Biology and Physiology and the McAllister Heart Institute, University of North Carolina at Chapel Hill, United States of America
| | - Brian K Buckley
- Department of Cell Biology and Physiology and the McAllister Heart Institute, University of North Carolina at Chapel Hill, United States of America
| | - Erika S Wittchen
- Department of Cell Biology and Physiology and the McAllister Heart Institute, University of North Carolina at Chapel Hill, United States of America
| | - Willa Y Ma
- Department of Cell Biology and Physiology and the McAllister Heart Institute, University of North Carolina at Chapel Hill, United States of America
| | - James E Faber
- Department of Cell Biology and Physiology and the McAllister Heart Institute, University of North Carolina at Chapel Hill, United States of America.
| |
Collapse
|
30
|
Klems A, van Rijssel J, Ramms AS, Wild R, Hammer J, Merkel M, Derenbach L, Préau L, Hinkel R, Suarez-Martinez I, Schulte-Merker S, Vidal R, Sauer S, Kivelä R, Alitalo K, Kupatt C, van Buul JD, le Noble F. The GEF Trio controls endothelial cell size and arterial remodeling downstream of Vegf signaling in both zebrafish and cell models. Nat Commun 2020; 11:5319. [PMID: 33087700 PMCID: PMC7578835 DOI: 10.1038/s41467-020-19008-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 09/22/2020] [Indexed: 02/07/2023] Open
Abstract
Arterial networks enlarge in response to increase in tissue metabolism to facilitate flow and nutrient delivery. Typically, the transition of a growing artery with a small diameter into a large caliber artery with a sizeable diameter occurs upon the blood flow driven change in number and shape of endothelial cells lining the arterial lumen. Here, using zebrafish embryos and endothelial cell models, we describe an alternative, flow independent model, involving enlargement of arterial endothelial cells, which results in the formation of large diameter arteries. Endothelial enlargement requires the GEF1 domain of the guanine nucleotide exchange factor Trio and activation of Rho-GTPases Rac1 and RhoG in the cell periphery, inducing F-actin cytoskeleton remodeling, myosin based tension at junction regions and focal adhesions. Activation of Trio in developing arteries in vivo involves precise titration of the Vegf signaling strength in the arterial wall, which is controlled by the soluble Vegf receptor Flt1. Arterial flow regulates artery diameter but other mechanisms may also affect this. Here, the authors show that the guanine nucleotide exchange factor Trio and GTPases Rac1 and RhoG, triggers F-actin remodeling in arterial endothelial cells, independent of flow, to enhance lumen diameter in zebrafish and cell models.
Collapse
Affiliation(s)
- Alina Klems
- Department of Cell and Developmental Biology, Institute of Zoology (ZOO), Karlsruhe Institute of Technology (KIT), Fritz Haber Weg 4, 76131, Karlsruhe, Germany
| | - Jos van Rijssel
- Molecular Cell Biology lab, Department Molecular and Cellular Hemostasis, Sanquin Research and Landsteiner Laboratory, Academic Medical Center at the University of Amsterdam, Plesmanlaan 125, 1066CX, Amsterdam, The Netherlands
| | - Anne S Ramms
- Department of Cell and Developmental Biology, Institute of Zoology (ZOO), Karlsruhe Institute of Technology (KIT), Fritz Haber Weg 4, 76131, Karlsruhe, Germany.,Institute for Biological and Chemical Systems-Biological Information Processing, Karlsruhe Institute of Technology (KIT), PO Box 3640, 76021, Karlsruhe, Germany
| | - Raphael Wild
- Department of Cell and Developmental Biology, Institute of Zoology (ZOO), Karlsruhe Institute of Technology (KIT), Fritz Haber Weg 4, 76131, Karlsruhe, Germany
| | - Julia Hammer
- Department of Cell and Developmental Biology, Institute of Zoology (ZOO), Karlsruhe Institute of Technology (KIT), Fritz Haber Weg 4, 76131, Karlsruhe, Germany
| | - Melanie Merkel
- Department of Cell and Developmental Biology, Institute of Zoology (ZOO), Karlsruhe Institute of Technology (KIT), Fritz Haber Weg 4, 76131, Karlsruhe, Germany
| | - Laura Derenbach
- Department of Cell and Developmental Biology, Institute of Zoology (ZOO), Karlsruhe Institute of Technology (KIT), Fritz Haber Weg 4, 76131, Karlsruhe, Germany
| | - Laetitia Préau
- Department of Cell and Developmental Biology, Institute of Zoology (ZOO), Karlsruhe Institute of Technology (KIT), Fritz Haber Weg 4, 76131, Karlsruhe, Germany
| | - Rabea Hinkel
- Laboratory Animal Science Unit, Leibnitz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Kellnerweg 4, 37077 Göttingen, Germany and DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany
| | - Irina Suarez-Martinez
- Institute of Cardiovascular Organogenesis and Regeneration WWU Münster, Münster, Germany & Faculty of Medicine, WWU Münster, Münster, Germany & Cells in Motion Cluster of Excellence, Münster, Münster, Germany
| | - Stefan Schulte-Merker
- Institute of Cardiovascular Organogenesis and Regeneration WWU Münster, Münster, Germany & Faculty of Medicine, WWU Münster, Münster, Germany & Cells in Motion Cluster of Excellence, Münster, Münster, Germany
| | - Ramon Vidal
- Max Delbrück Center for Molecular Medicine (MDC), Berlin Institute of Medical Systems Biology & Berlin Institute of Health, Robert Rössle Strasse 10, 13092, Berlin, Germany
| | - Sascha Sauer
- Max Delbrück Center for Molecular Medicine (MDC), Berlin Institute of Medical Systems Biology & Berlin Institute of Health, Robert Rössle Strasse 10, 13092, Berlin, Germany
| | - Riikka Kivelä
- Stem Cells and Metabolism Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, and Wihuri Research Institute, Helsinki, Finland
| | - Kari Alitalo
- Translational Cancer Medicine Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, and Wihuri Research Institute, Helsinki, Finland
| | - Christian Kupatt
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, TUM Munich, Germany, and DZHK, (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Jaap D van Buul
- Molecular Cell Biology lab, Department Molecular and Cellular Hemostasis, Sanquin Research and Landsteiner Laboratory, Academic Medical Center at the University of Amsterdam, Plesmanlaan 125, 1066CX, Amsterdam, The Netherlands.,Leeuwenhoek Centre for Advanced Microscopy, section Molecular Cytology at Swammerdam Institute for Life Sciences at University of Amsterdam, Amsterdam, The Netherlands
| | - Ferdinand le Noble
- Department of Cell and Developmental Biology, Institute of Zoology (ZOO), Karlsruhe Institute of Technology (KIT), Fritz Haber Weg 4, 76131, Karlsruhe, Germany. .,Institute for Biological and Chemical Systems-Biological Information Processing, Karlsruhe Institute of Technology (KIT), PO Box 3640, 76021, Karlsruhe, Germany. .,Institute of Experimental Cardiology, University of Heidelberg, Heidelberg Germany and DZHK (German Center for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
| |
Collapse
|
31
|
Ravindran AV, Killingsworth MC, Bhaskar S. Cerebral collaterals in acute ischaemia: Implications for acute ischaemic stroke patients receiving reperfusion therapy. Eur J Neurosci 2020; 53:1238-1261. [PMID: 32871623 DOI: 10.1111/ejn.14955] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/20/2020] [Accepted: 08/22/2020] [Indexed: 12/21/2022]
Abstract
The cerebral collaterals play an important role in penumbral tissue sustenance after an acute ischaemic stroke. Recent studies have demonstrated the potential role of collaterals in the selection of acute ischaemic stroke patients eligible for reperfusion therapy. However, the understanding of the significance and evidence around the role of collateral status in predicting outcomes in acute ischaemic stroke patients treated with reperfusion therapy is still unclear. Moreover, the use of pre-treatment collaterals in patient selection and prognosis is relatively underappreciated in clinical settings. A focused review of the literature was performed on the various methods of collateral evaluation and the role of collateral status in acute ischaemic stroke patients receiving reperfusion therapy. We discuss the methods of evaluating pre-treatment collaterals in clinical settings. The patient selection based on collateral status as well as the prognostic and therapeutic value of collaterals in acute ischaemic stroke, in settings of intravenous thrombolysis or endovascular therapy alone, and bridge therapy, are summarized. Recommendations for future research and possible pharmacological intervention strategies aimed at collateral enhancement are also discussed. Collaterals may play an important role in identifying acute ischaemic stroke patients who are likely to benefit from endovascular treatment in an extended time window. Future neuroscientific efforts to better improve our understanding of the role of collaterals in acute ischaemia as well as clinical studies to delineate its role in patient selection and acute stroke prognosis are warranted.
Collapse
Affiliation(s)
- Abina Vishni Ravindran
- South Western Sydney Clinical School, University of New South Wales (UNSW), Sydney, NSW, Australia.,Neurovascular Imaging Laboratory, Clinical Sciences Stream, Ingham Institute for Applied Medical Research, Sydney, NSW, Australia.,Thrombolysis and Endovascular WorkFLOw Network (TEFLON), Sydney, NSW, Australia
| | - Murray C Killingsworth
- South Western Sydney Clinical School, University of New South Wales (UNSW), Sydney, NSW, Australia.,NSW Brain Clot Bank, NSW Health Statewide Biobank and NSW Health Pathology, Sydney, NSW, Australia.,Correlative Microscopy Facility, Ingham Institute for Applied Medical Research and Department of Anatomical Pathology, NSW Health Pathology and Liverpool Hospital, Liverpool, NSW, Australia
| | - Sonu Bhaskar
- South Western Sydney Clinical School, University of New South Wales (UNSW), Sydney, NSW, Australia.,Department of Neurology & Neurophysiology, Liverpool Hospital & South West Sydney Local Health District (SWSLHD), Sydney, NSW, Australia.,Neurovascular Imaging Laboratory, Clinical Sciences Stream, Ingham Institute for Applied Medical Research, Sydney, NSW, Australia.,Stroke & Neurology Research Group, Ingham Institute for Applied Medical Research, Sydney, NSW, Australia.,NSW Brain Clot Bank, NSW Health Statewide Biobank and NSW Health Pathology, Sydney, NSW, Australia.,Thrombolysis and Endovascular WorkFLOw Network (TEFLON), Sydney, NSW, Australia
| |
Collapse
|
32
|
Zhang H, Rzechorzek W, Aghajanian A, Faber JE. Hypoxia induces de novo formation of cerebral collaterals and lessens the severity of ischemic stroke. J Cereb Blood Flow Metab 2020; 40:1806-1822. [PMID: 32423327 PMCID: PMC7430105 DOI: 10.1177/0271678x20924107] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Pial collaterals provide protection in stroke. Evidence suggests their formation late during gestation (collaterogenesis) is driven by reduced oxygen levels in the cerebral watersheds. The purpose of this study was to determine if collaterogenesis can be re-activated in the adult to induce formation of additional collaterals ("neo-collateral formation", NCF). Mice were gradually acclimated to reduced inspired oxygen (FIO2) and maintained at 12, 10, 8.5 or 7% for two-to-eight weeks. Hypoxemia induced "dose"-dependent NCF and remodeling of native collaterals, and decreased infarct volume after permanent MCA occlusion. In contrast, no formation occurred of addition collateral-like intra-tree anastomoses, PComs, or branches within the MCA tree. Hypoxic NCF, remodeling and infarct protection were durable, i.e. retained for at least six weeks after return to normoxia. Hypoxia increased expression of Hif2α, Vegfa, Rabep2, Angpt2, Tie2 and Cxcr4. Neo-collateral formation was abolished in mice lacking Rabep2, a novel gene involved in VEGFA→Flk1 signaling and required for formation of collaterals during development, and inhibited by knockdown of Vegfa, Flk1 and Cxcr4. Rabep2-dependent NCF was also induced by permanent MCA occlusion. This is the first report that hypoxia induces new pial collaterals to form. Hypoxia- and occlusion-induced neo-collateral formation provide models to study collaterogenesis in the adult.
Collapse
Affiliation(s)
- Hua Zhang
- Department of Cell Biology and Physiology, McAllister Heart Institute, Curriculum in Neurobiology, University of North Carolina at Chapel Hill, NC, USA
| | - Wojciech Rzechorzek
- Department of Cell Biology and Physiology, McAllister Heart Institute, Curriculum in Neurobiology, University of North Carolina at Chapel Hill, NC, USA
| | - Amir Aghajanian
- Department of Cell Biology and Physiology, McAllister Heart Institute, Curriculum in Neurobiology, University of North Carolina at Chapel Hill, NC, USA
| | - James E Faber
- Department of Cell Biology and Physiology, McAllister Heart Institute, Curriculum in Neurobiology, University of North Carolina at Chapel Hill, NC, USA
| |
Collapse
|
33
|
Recruitment and maturation of the coronary collateral circulation: Current understanding and perspectives in arteriogenesis. Microvasc Res 2020; 132:104058. [PMID: 32798552 DOI: 10.1016/j.mvr.2020.104058] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 06/09/2020] [Accepted: 08/11/2020] [Indexed: 12/13/2022]
Abstract
The coronary collateral circulation is a rich anastomotic network of primitive vessels which have the ability to augment in size and function through the process of arteriogenesis. In this review, we evaluate the current understandings of the molecular and cellular mechanisms by which this process occurs, specifically focussing on elevated fluid shear stress (FSS), inflammation, the redox state and gene expression along with the integrative, parallel and simultaneous process by which this occurs. The initiating step of arteriogenesis occurs following occlusion of an epicardial coronary artery, with an increase in FSS detected by mechanoreceptors within the endothelium. This must occur within a 'redox window' where an equilibrium of oxidative and reductive factors are present. These factors initially result in an inflammatory milieu, mediated by neutrophils as well as lymphocytes, with resultant activation of a number of downstream molecular pathways resulting in increased expression of proteins involved in monocyte attraction and adherence; namely vascular cell adhesion molecule 1 (VCAM-1), monocyte chemoattractant protein 1 (MCP-1) and transforming growth factor beta (TGF-β). Once monocytes and other inflammatory cells adhere to the endothelium they enter the extracellular matrix and differentiate into macrophages in an effort to create a favourable environment for vessel growth and development. Activated macrophages secrete inflammatory cytokines such as tumour necrosis factor-α (TNF-α), growth factors such as fibroblast growth factor-2 (FGF-2) and matrix metalloproteinases. Finally, vascular smooth muscle cells proliferate and switch to a contractile phenotype, resulting in an increased diameter and functionality of the collateral vessel, thereby allowing improved perfusion of the distal myocardium subtended by the occluded vessel. This simultaneously reduces FSS within the collateral vessel, inhibiting further vessel growth.
Collapse
|
34
|
Ma J, Ma Y, Shuaib A, Winship IR. Impaired Collateral Flow in Pial Arterioles of Aged Rats During Ischemic Stroke. Transl Stroke Res 2020; 11:243-253. [PMID: 31203565 PMCID: PMC7067739 DOI: 10.1007/s12975-019-00710-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 05/02/2019] [Accepted: 06/05/2019] [Indexed: 02/05/2023]
Abstract
Cerebral collateral circulation and age are critical factors in determining outcome from acute ischemic stroke. Aging may lead to rarefaction of cerebral collaterals, and thereby accelerate ischemic injury by reducing penumbral blood flow. Dynamic changes in pial collaterals after onset of cerebral ischemia may vary with age but have not been extensively studied. Here, laser speckle contrast imaging (LSCI) and two-photon laser scanning microscopy (TPLSM) were combined to monitor cerebral pial collaterals between the anterior cerebral artery (ACA) and the middle cerebral artery (MCA) in young adult and aged male Sprague Dawley rats during distal middle cerebral artery occlusion (dMCAo). Histological analysis showed that aged rats had significantly greater volumes of ischemic damage than young rats. LSCI showed that cerebral collateral perfusion declined over time after stroke in aged and young rats, and that this decline was significantly greater in aged rats. TPLSM demonstrated that pial arterioles narrowed faster after dMCAo in aged rats compared to young adult rats. Notably, while arteriole vessel narrowing was comparable 4.5 h after ischemic onset in aged and young adult rats, red blood cell velocity was stable in young adults but declined over time in aged rats. Overall, red blood cell flux through pial arterioles was significantly reduced at all time-points after 90 min post-dMCAo in aged rats relative to young adult rats. Thus, collateral failure is more severe in aged rats with significantly impaired pial collateral dynamics (reduced diameter, red blood cell velocity, and red blood cell flux) relative to young adult rats.
Collapse
Affiliation(s)
- Junqiang Ma
- Neurochemical Research Unit, Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, 12-127 Clinical Sciences Building, Edmonton, AB, T6G 2R3, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Yonglie Ma
- Neurochemical Research Unit, Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, 12-127 Clinical Sciences Building, Edmonton, AB, T6G 2R3, Canada
| | - Ashfaq Shuaib
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Division of Neurology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Ian R Winship
- Neurochemical Research Unit, Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, 12-127 Clinical Sciences Building, Edmonton, AB, T6G 2R3, Canada.
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
35
|
Marushima A, Nieminen M, Kremenetskaia I, Gianni-Barrera R, Woitzik J, von Degenfeld G, Banfi A, Vajkoczy P, Hecht N. Balanced single-vector co-delivery of VEGF/PDGF-BB improves functional collateralization in chronic cerebral ischemia. J Cereb Blood Flow Metab 2020; 40:404-419. [PMID: 30621518 PMCID: PMC7370608 DOI: 10.1177/0271678x18818298] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The myoblast-mediated delivery of angiogenic genes represents a cell-based approach for targeted induction of therapeutic collateralization. Here, we tested the superiority of myoblast-mediated co-delivery of vascular endothelial growth factor-A (VEGF) together with platelet-derived growth factor-BB (PDGF-BB) on transpial collateralization of an indirect encephalomyosynangiosis (EMS) in a model of chronic cerebral ischemia. Mouse myoblasts expressing a reporter gene alone (empty vector), VEGF, PDGF-BB or VEGF and PDGF-BB through a single bi-cistronic vector (VIP) were implanted into the temporalis muscle of an EMS following permanent ipsilateral internal carotid artery occlusion in adult, male C57BL/6N mice. Over 84 days, myoblast engraftment and gene product expression, hemodynamic impairment, transpial collateralization, angiogenesis, pericyte recruitment and post-ischemic neuroprotection were assessed. By day 42, animals that received PDGF-BB in combination with VEGF (VIP) showed superior hemodynamic recovery, EMS collateralization and ischemic protection with improved pericyte recruitment around the parenchymal vessels and EMS collaterals. Also, supplementation of PDGF-BB resulted in a striking astrocytic activation with intrinsic VEGF mobilization in the cortex below the EMS. Our findings suggest that EMS surgery together with myoblast-mediated co-delivery of VEGF/PDGF-BB may have the potential to serve as a novel treatment strategy for augmentation of collateral flow in the chronically hypoperfused brain.
Collapse
Affiliation(s)
- Aiki Marushima
- Department of Neurosurgery and Center for Stroke research Berlin (CSB), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Melina Nieminen
- Department of Neurosurgery and Center for Stroke research Berlin (CSB), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Irina Kremenetskaia
- Department of Neurosurgery and Center for Stroke research Berlin (CSB), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Roberto Gianni-Barrera
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Johannes Woitzik
- Department of Neurosurgery and Center for Stroke research Berlin (CSB), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | - Andrea Banfi
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Peter Vajkoczy
- Department of Neurosurgery and Center for Stroke research Berlin (CSB), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Nils Hecht
- Department of Neurosurgery and Center for Stroke research Berlin (CSB), Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
36
|
Isoform-Specific Roles of ERK1 and ERK2 in Arteriogenesis. Cells 2019; 9:cells9010038. [PMID: 31877781 PMCID: PMC7017123 DOI: 10.3390/cells9010038] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/10/2019] [Accepted: 12/18/2019] [Indexed: 12/14/2022] Open
Abstract
Despite the clinical importance of arteriogenesis, this biological process is poorly understood. ERK1 and ERK2 are key components of a major intracellular signaling pathway activated by vascular endothelial growth (VEGF) and FGF2, growth factors critical to arteriogenesis. To investigate the specific role of each ERK isoform in arteriogenesis, we used mice with a global Erk1 knockout as well as Erk1 and Erk2 floxed mice to delete Erk1 or Erk2 in endothelial cells, macrophages, and smooth muscle cells. We found that ERK1 controls macrophage infiltration following an ischemic event. Loss of ERK1 in endothelial cells and macrophages induced an excessive macrophage infiltration leading to an increased but poorly functional arteriogenesis. Loss of ERK2 in endothelial cells leads to a decreased arteriogenesis due to decreased endothelial cell proliferation and a reduced eNOS expression. These findings show for the first time that isoform-specific roles of ERK1 and ERK2 in the control of arteriogenesis.
Collapse
|
37
|
Transient versus Permanent MCA Occlusion in Mice Genetically Modified to Have Good versus Poor Collaterals. ACTA ACUST UNITED AC 2019; 4. [PMID: 31840083 PMCID: PMC6910253 DOI: 10.20900/mo.20190024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Collateral-dependent blood flow is capable of significantly lessening the severity of stroke. Unfortunately, collateral flow varies widely in patients for reasons that remain unclear. Studies in mice have shown that the number and diameter of cerebral collaterals vary widely due primarily to polymorphisms in genes, e.g., Rabep2, involved in their formation during development. However, understanding how variation in collateral abundance affects stroke progression has been hampered by lack of a method to reversibly ligate the distal middle cerebral artery (MCAO) in mice. Here we present a method and examine infarct volume 24 h after transient (tMCAO, 90 min) versus permanent occlusion (pMCAO) in mice with good versus poor collaterals. Wildtype C57BL/6 mice (have abundant collaterals) sustained small infarctions following tMCAO that increased 2.1-fold after pMCAO, reflecting significant penumbra present at 90 min. Mutant C57BL/6 mice lacking Rabep2 (have reduced collaterals) sustained a 4-fold increase in infarct volume over WT following tMCAO and a smaller additional increase (0.4-fold) after pMCAO, reflecting reduced penumbra. Wildtype BALB/cBy (have a deficient Rabep2 variant and poor collaterals) had large infarctions following tMCAO that increased less (0.6-fold) than the above wildtype C57BL/6 mice following pMCAO. Mutant BALB/cBy mice (have deficient Rabep2 replaced with the C57BL/6 variant thus increased collaterals) sustained smaller infarctions after tMCAO. However, unlike C57BL/6 versus Rabep2 mice, penumbra was not increased since infarct volume increased only 0.3-fold following pMCAO. These findings present a murine model of tMCAO and demonstrate that neuroprotective mechanisms, in addition to collaterals, also vary with genetic background and affect the evolution of stroke.
Collapse
|
38
|
The role of endothelial cells in cystic fibrosis. J Cyst Fibros 2019; 18:752-761. [DOI: 10.1016/j.jcf.2019.07.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 06/18/2019] [Accepted: 07/23/2019] [Indexed: 12/22/2022]
|
39
|
Fusconi M, Attanasio G, Capitani F, Di Porto E, Diacinti D, Musy I, Ralli M, Ralli G, Greco A, de Vincentiis M, Colonnese C. Is there a relation between sudden sensorineural hearing loss and white matter lesions? Eur Arch Otorhinolaryngol 2019; 276:3043-3049. [PMID: 31410545 DOI: 10.1007/s00405-019-05593-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 08/01/2019] [Indexed: 12/22/2022]
Abstract
PURPOSE Sudden sensorineural hearing loss (SSNHL) has similarities to conditions with vascular etiologies such as myocardial infarction and cerebral stroke. Thus, it could be considered as an early sign of a vascular disease and not only a specific local condition. Chronic hypoperfusion in the brain districts leads to a chronic ischemic damage, called cerebral small vessel disease (CSVD), detectable with brain magnetic resonance imaging (MRI). METHODS The authors used CSVD to establish the presence of vascular risk factors in individuals with SSNHL and used the Fazekas score scale to classify them. RESULTS Our study showed that individuals with SSNHL aged between 48 and 60 years have 26% more probability to have a Fazekas score higher than 1 compared to the general population. Individuals younger than 28 years showed a statistically significant negative correlation to have a Fazekas score higher than 0. The higher is the Fazekas score, the less is the probability of hearing recovery. The medium hearing-recovery probability is 46%. This decreases by 16% for every increase of score in the Fazekas scale. In the present study, the recovery probability decreased from 80% in individuals younger than 48 years with a score of 0 to 14% in individuals with a Fazekas scores of 3 and 4. CONCLUSIONS The authors assessed a higher prevalence of CSVD compared to the general population in patients aged between 48 and 60 years with SSNHL. Moreover, they assessed that the presence of CSVD is related to a decreased probability of recovery, as it has already been demonstrated for stroke.
Collapse
Affiliation(s)
- Massimo Fusconi
- Department of Sense Organs, Sapienza University of Rome, Rome, Italy
| | | | - Flavia Capitani
- Clinic for Ear, Nose and Throat Medicine, Uniklinik of Tuebingen, Albstrasse 93, 70597, Stuttgart, Germany.
| | - Edoardo Di Porto
- Department of Economics and Statistics, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, Naples, Italy
| | - Davide Diacinti
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, University Sapienza, Rome, Italy
| | - Isotta Musy
- Department of Sense Organs, Sapienza University of Rome, Rome, Italy
| | - Massimo Ralli
- Department of Sense Organs, Sapienza University of Rome, Rome, Italy
| | - Giovanni Ralli
- Department of Sense Organs, Sapienza University of Rome, Rome, Italy
| | - Antonio Greco
- Department of Sense Organs, Sapienza University of Rome, Rome, Italy
| | - Marco de Vincentiis
- Department of Oral and Maxillofacial Sciences, Sapienza University of Rome, Rome, Italy
| | - Claudio Colonnese
- Department of Neurology and Psichiatry, Neuroradiology Section, University Sapienza, Rome, Italy.,IRCCS Neuromed, Pozzilli, IS, Italy
| |
Collapse
|
40
|
Collateral Vessels Have Unique Endothelial and Smooth Muscle Cell Phenotypes. Int J Mol Sci 2019; 20:ijms20153608. [PMID: 31344780 PMCID: PMC6695737 DOI: 10.3390/ijms20153608] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/11/2019] [Accepted: 07/19/2019] [Indexed: 12/15/2022] Open
Abstract
Collaterals are unique blood vessels present in the microcirculation of most tissues that, by cross-connecting a small fraction of the outer branches of adjacent arterial trees, provide alternate routes of perfusion. However, collaterals are especially susceptible to rarefaction caused by aging, other vascular risk factors, and mouse models of Alzheimer’s disease—a vulnerability attributed to the disturbed hemodynamic environment in the watershed regions where they reside. We examined the hypothesis that endothelial and smooth muscle cells (ECs and SMCs, respectively) of collaterals have specializations, distinct from those of similarly-sized nearby distal-most arterioles (DMAs) that maintain collateral integrity despite their continuous exposure to low and oscillatory/disturbed shear stress, high wall stress, and low blood oxygen. Examination of mouse brain revealed the following: Unlike the pro-inflammatory cobble-stoned morphology of ECs exposed to low/oscillatory shear stress elsewhere in the vasculature, collateral ECs are aligned with the vessel axis. Primary cilia, which sense shear stress, are present, unexpectedly, on ECs of collaterals and DMAs but are less abundant on collaterals. Unlike DMAs, collaterals are continuously invested with SMCs, have increased expression of Pycard, Ki67, Pdgfb, Angpt2, Dll4, Ephrinb2, and eNOS, and maintain expression of Klf2/4. Collaterals lack tortuosity when first formed during development, but tortuosity becomes evident within days after birth, progresses through middle age, and then declines—results consistent with the concept that collateral wall cells have a higher turnover rate than DMAs that favors proliferative senescence and collateral rarefaction. In conclusion, endothelial and SMCs of collaterals have morphologic and functional differences from those of nearby similarly sized arterioles. Future studies are required to determine if they represent specializations that counterbalance the disturbed hemodynamic, pro-inflammatory, and pro-proliferative environment in which collaterals reside and thus mitigate their risk factor-induced rarefaction.
Collapse
|
41
|
Liu H, Wang H, Cheng D, Wang Q, Pei Z, Zhu N, Fang W, Yu Q. Potential role of a disintegrin and metalloproteinase-17 (ADAM17) in age-associated ventricular remodeling of rats. RSC Adv 2019; 9:14321-14330. [PMID: 35519346 PMCID: PMC9064162 DOI: 10.1039/c9ra01190k] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 04/23/2019] [Indexed: 01/03/2023] Open
Abstract
Excessive tumor necrosis factor-α (TNF-α) could enhance cell death and aggravate left ventricular remodeling and myocardial dysfunction. A disintegrin and metalloproteinase-17 (ADAM17), an important maturation regulator of TNF-α, might be involved in the aging-associated ventricular remodeling. The present study observed myocardial ADAM17 expression in young and aged rats and explored the association between cardiac structure/function and expression of ADAM17 in 6 month-old (n = 10, young group) and 24 month-old SD rats (n = 10, old group). The body, heart weight and heart weight/body weight ratio of rats in the old group were all significantly increased compared to that in the young group (P < 0.05). The left ventricular systolic end-diameter and end-diastolic diameters were significantly enlarged in the old group compared to the young group (P < 0.05), while the systolic function index including the left ventricular ejection fraction and left ventricular fractional shortening were similar between the two groups. The peak mitral flow velocity (E)/peak mitral annulus velocity (E') ratio was significantly higher in the old group than in the young group (P < 0.05). Histological examination showed more damage of cardiomyocytes, interstitial collagen deposition and inflammatory cell infiltration in the old group. Immunohistochemistry examination showed that myocardial TNF-α expression was mainly located in cardiomyocytes and was significantly higher in the old group than in the young group (P < 0.05). The protein expression of myocardial ADAM17 detected by western blot was significantly higher in the old group than in the young group (P < 0.05), while TIMP-3 expression was similar between the two groups. The present study suggested that ADAM17 and inflammation might play an important role in aging-related myocardial remodeling through regulating TNF-α.
Collapse
Affiliation(s)
- Hainiang Liu
- Department of Cardiology, Affiliated Zhongshan Hospital of Dalian University Dalian 116001 China +86-411-62893555 +86-411-62887018.,Medical College, Dalian University Dalian 116622 China
| | - Haoren Wang
- Department of Cardiology, Affiliated Zhongshan Hospital of Dalian University Dalian 116001 China +86-411-62893555 +86-411-62887018
| | - Dong Cheng
- Department of Cardiology, Affiliated Zhongshan Hospital of Dalian University Dalian 116001 China +86-411-62893555 +86-411-62887018.,Medical College, Dalian University Dalian 116622 China
| | - Qinfu Wang
- Life Engineering College, Dalian University Dalian 116622 China
| | - Zuowei Pei
- Department of Cardiology, Affiliated Zhongshan Hospital of Dalian University Dalian 116001 China +86-411-62893555 +86-411-62887018
| | - Ning Zhu
- Department of Cardiology, The Second Affiliated Hospital of Dalian Medical University Dalian 116023 China
| | - Weiyi Fang
- Department of Cardiology, Shanghai Chest Hospital Shanghai 200030 China
| | - Qin Yu
- Department of Cardiology, Affiliated Zhongshan Hospital of Dalian University Dalian 116001 China +86-411-62893555 +86-411-62887018
| |
Collapse
|
42
|
Genetic and Environmental Contributions to Variation in the Posterior Communicating Collaterals of the Circle of Willis. Transl Stroke Res 2019; 10:189-203. [PMID: 29589286 DOI: 10.1007/s12975-018-0626-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 03/14/2018] [Accepted: 03/16/2018] [Indexed: 02/07/2023]
Abstract
Variation in blood flow mediated by the posterior communicating collateral arteries (PComs) contributes to variation in the severity of tissue injury in obstructive disease. Evidence in animals and humans indicates that differences in the extent of PComs, i.e., their anatomic lumen diameter and whether they are present bilaterally, unilaterally, or absent, are a major factor. These differences arise during development since they are present at birth. However, the causal mechanisms are unknown. We used angiography after maximal dilation to examine involvement of genetic, environmental, and stochastic factors. The extent of PComs varied widely among seven genetically diverse strains of mice. Like pial collaterals in the microcirculation, aging and hypertension reduced PCom diameter, while in contrast, obesity, hyperlipidemia, metabolic syndrome, and diabetes mellitus had no effect. Naturally occurring intrauterine growth restriction had no effect on extent of PCom or pial collaterals in the adult. The number and diameter of PComs evidenced much larger apparent stochastic-dependent variation than pial collaterals. In addition, both PComs underwent flow-mediated outward remodeling after unilateral permanent MCA occlusion that varied with genetic background and was greater on the ipsilesional side. These findings indicate that variation in the number and diameter of PCom collateral arteries arises from stochastic factors and naturally occurring genetic variants that differ from those that cause variation in pial collateral arterioles. Environmental factors also contribute: aging and hypertension reduce PCom diameter. Our results suggest possible sources of variation of PComs in humans and provide information relevant when studying mouse models of occlusive cerebrovascular disease.
Collapse
|
43
|
Piedade GS, Schirmer CM, Goren O, Zhang H, Aghajanian A, Faber JE, Griessenauer CJ. Cerebral Collateral Circulation: A Review in the Context of Ischemic Stroke and Mechanical Thrombectomy. World Neurosurg 2019; 122:33-42. [PMID: 30342266 DOI: 10.1016/j.wneu.2018.10.066] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 10/06/2018] [Accepted: 10/08/2018] [Indexed: 01/28/2023]
Abstract
The pial (leptomenigeal) collateral circulation is a key determinant of functional outcome after mechanical thrombectomy after large-vessel ischemic stroke. Patients with good collateral blood flow benefit up to 24 hours after stroke onset, whereas those with poor collateral flow evidence less or no benefit. However, clues to why collateral flow varies so widely among patients have remained elusive. Recent findings in animal studies, which are currently being tested for confirmation in humans, have found that naturally occurring variants of a novel "collateral gene," Rabep2, result in large differences in the extent of anatomic collaterals and thus blood flow and infarct size in mice after stroke. The comprehension of collagerogenesis in humans and the evaluation of collateral status could aid in identifying patients who will benefit not only from mechanical thrombectomy in the extended time window but also from any reperfusion strategy. We performed a literature review focused on radiographic, clinical, and genetic aspects of the collateral circulation.
Collapse
Affiliation(s)
- Guilherme Santos Piedade
- Department of Neurosurgery, Geisinger, Pennsylvania, USA; Department of Neurosurgery, University of Düsseldorf, Düsseldorf, Germany
| | | | - Oded Goren
- Department of Neurosurgery, Geisinger, Pennsylvania, USA
| | - Hua Zhang
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Amir Aghajanian
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - James E Faber
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Christoph J Griessenauer
- Department of Neurosurgery, Geisinger, Pennsylvania, USA; Research Institute of Neurointervention, Paracelsus Medical University, Salzburg, Austria; Department of Neurosurgery, Paracelsus Medical University, Salzburg, Austria.
| |
Collapse
|
44
|
Mouse models of Alzheimer's disease cause rarefaction of pial collaterals and increased severity of ischemic stroke. Angiogenesis 2019; 22:263-279. [PMID: 30519973 PMCID: PMC6475514 DOI: 10.1007/s10456-018-9655-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 11/20/2018] [Indexed: 01/26/2023]
Abstract
Vascular dysfunction contributes to the progression and severity of Alzheimer's disease (AD). Patients with AD also sustain larger infarctions after ischemic stroke; however, the responsible mechanisms are unknown. Pial collaterals are the primary source of protection in stroke. Unfortunately, natural aging and other vascular risk factors cause a decline in collateral number and diameter (rarefaction) and an increase in stroke severity. Herein, we tested the hypothesis that AD accelerates age-induced collateral rarefaction and examined potential underlying mechanisms. Triple and double transgenic mouse models of AD both sustained collateral rarefaction by 8 months of age, well before the onset of rarefaction caused by aging alone (16 months of age). Rarefaction, which did not progress further at 18 months of age, was accompanied by a twofold increase in infarct volume after MCA occlusion. AD did not induce rarefaction of similarly sized pial arterioles or penetrating arterioles. Rarefaction was minimal and occurred only at 18 months of age in a parenchymal vascular amyloid-beta model of AD. Rarefaction was not associated with amyloid-beta deposition on collaterals or pial arteries, nor was plaque burden or CD11b+ cell density greater in brain underlying the collateral zones versus elsewhere. However, rarefaction was accompanied by increased markers of oxidative stress, inflammation, and aging of collateral endothelial and mural cells. Moreover, rarefaction was lessened by deletion of CX3CR1 and prevented by overexpression of eNOS. These findings demonstrate that mouse models of AD promote rarefaction of pial collaterals and implicate inflammation-induced accelerated aging of collateral wall cells. Strategies that reduce vascular inflammation and/or increase nitric oxide may preserve collateral function.
Collapse
|
45
|
Simons KH, Aref Z, Peters HAB, Welten SP, Nossent AY, Jukema JW, Hamming JF, Arens R, de Vries MR, Quax PHA. The role of CD27-CD70-mediated T cell co-stimulation in vasculogenesis, arteriogenesis and angiogenesis. Int J Cardiol 2018; 260:184-190. [PMID: 29622436 DOI: 10.1016/j.ijcard.2018.02.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 01/25/2018] [Accepted: 02/02/2018] [Indexed: 01/04/2023]
Abstract
BACKGROUND T cells have a distinctive role in neovascularization, which consists of arteriogenesis and angiogenesis under pathological conditions and vasculogenesis under physiological conditions. However, the role of co-stimulation in T cell activation in neovascularization has yet to be established. The aim of this study was to investigate the role T cell co-stimulation and inhibition in angiogenesis, arteriogenesis and vasculogenesis. METHODS AND RESULTS Hind limb ischemia was induced by double ligation of the left femoral artery in mice and blood flow recovery was measured with Laser Doppler Perfusion Imaging in control, CD70-/-, CD80/86-/-, CD70/80/86-/- and CTLA4+/- mice. Blood flow recovery was significantly impaired in mice lacking CD70 compared to control mice, but was similar in CD80/86-/-, CTLA4+/- and control mice. Mice lacking CD70 showed impaired vasculogenesis, since the number of pre-existing collaterals was reduced as observed in the pia mater compared to control mice. In vitro an impaired capability of vascular smooth muscle cells (VSMC) to activate T cells was observed in VSMC lacking CD70. Furthermore, CD70-/-, CD80/86-/- and CD70/80/86-/- mice showed reduced angiogenesis in the soleus muscle 10 days after ligation. Arteriogenesis was also decreased in CD70-/- compared to control mice 10 and 28 days after surgery. CONCLUSIONS The present study is the first to describe an important role for T cell activation via co-stimulation in angiogenesis, arteriogenesis and vasculogenesis, where the CD27-CD70 T cell co-stimulation pathway appears to be the most important co-stimulation pathway in pre-existing collateral formation and post-ischemic blood flow recovery, by arteriogenesis and angiogenesis.
Collapse
Affiliation(s)
- K H Simons
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands; Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Z Aref
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands; Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - H A B Peters
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands; Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - S P Welten
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - A Y Nossent
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands; Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - J W Jukema
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands; Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - J F Hamming
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - R Arens
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - M R de Vries
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands; Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - P H A Quax
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands; Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
46
|
Matei N, Camara R, Zhang JH. Emerging mechanisms and novel applications of hydrogen gas therapy. Med Gas Res 2018; 8:98-102. [PMID: 30319764 PMCID: PMC6178641 DOI: 10.4103/2045-9912.239959] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 08/03/2018] [Indexed: 12/13/2022] Open
Abstract
Clinical and pre-clinical studies have reported a broad range of applications for hydrogen gas therapy. Classically, conventional antioxidant therapy is limited because it neutralizes both the detrimental and protective effects of reactive oxygen species. As a weak reducing agent, hydrogen gas avoids this paradox by reacting with strong oxidants while leaving other beneficial oxidants reactive. This review gathers a promising list of hydrogen gas applications that merit further mechanistic investigation and additional therapeutic trials. Reports support the ability of hydrogen gas to downregulate the expression of pro-inflammatory cytokines and pro-apoptotic factors. Mechanistically, hydrogen gas has been shown to downregulate miR-9 and miR-21, while upregulating miR-199 to reduce inflammatory injury. In angiogenic pathways, hydrogen's inhibition of cyclic guanosine monophosphate-degrading phosphodiesterase led to higher levels of cyclic guanosine monophosphate, activation of protein kinase, and angiogenesis; next, as hydrogen gas increased the levels of intracellular calcium, stimulated vascular endothelial growth factor increased nitric oxide production. In conjunction, hydrogen gas opened adenosine triphosphate-sensitive potassium channel channels, which activate downstream mitogen-activated protein kinase pathways. Growing molecular mechanisms have discovered a plethora of downstream targets for hydrogen gas therapy that include autophagy (via the adenosine 5’-monophosphate-activated protein kinase/mammalian target of rapamycin pathway), histone modification, mitochondrial unfolded protein response, acute oxidative stress after exercise, and oxidative stress secondary to aging. In conclusion, evolving research has discovered novel molecular connections that will continue to widen applications for hydrogen therapy.
Collapse
Affiliation(s)
- Nathanael Matei
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Richard Camara
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA.,Department of Anesthesiology, Loma Linda University, Loma Linda, CA, USA.,Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
47
|
Advanced Neuroimaging of Acute Ischemic Stroke: Penumbra and Collateral Assessment. Neuroimaging Clin N Am 2018; 28:585-597. [PMID: 30322595 DOI: 10.1016/j.nic.2018.06.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Acute ischemic stroke (AIS) occurs when there is a sudden loss in cerebral blood flow due to embolic or thromboembolic occlusion of a cerebral or cervical artery. Patients with AIS require emergent neuroimaging to guide treatment, which includes intravenous thrombolysis and endovascular mechanical thrombectomy (EMT). Recent advances in AIS treatment by EMT has been driven in part by advances in computed tomography (CT) and MR imaging neuroimaging evaluation of ischemic penumbra and pial collateral vessels. The authors review advanced noninvasive brain imaging by CT and MR imaging for the evaluation of AIS focusing on penumbral and collateral imaging.
Collapse
|
48
|
Liebeskind DS. Mapping the collaterome for precision cerebrovascular health: Theranostics in the continuum of stroke and dementia. J Cereb Blood Flow Metab 2018; 38:1449-1460. [PMID: 28555527 PMCID: PMC6125977 DOI: 10.1177/0271678x17711625] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 04/04/2017] [Accepted: 04/29/2017] [Indexed: 01/10/2023]
Abstract
Precision cerebrovascular health or individualized long-term preservation of the brain and associated blood vessels, is predicated on understanding, diagnosing, and tailoring therapies for people at risk of ischemic injury associated with stroke and vascular dementia. The associated imaging patterns are sculpted by the protective effect of the collaterome, the innate compensatory ability of the brain and vasculature to offset hypoperfusion when antegrade or normal arterial inflow pathways are compromised. Theranostics or rational and synchronous use of diagnostic studies in tandem with specific therapies to optimally guide patient outcomes in ischemic brain disorders may capitalize on the pivotal role of the collaterome. Understanding the functional impact of the collaterome across populations of individuals would advance translational science on the brain, while questions with immediate clinical implications may be prioritized. Big data and systematic analyses are necessary to develop normative standards, multimodal imaging atlases, and delineation of specific patterns to guide clinical management. Large-scale, systematic imaging analyses of the collaterome provide a platform for translational work on cerebral collateral circulation and hemodynamics and a theranostic framework with direct clinical implications. This article frames incipient research objectives to guide precision stroke medicine in coming years, building upon the collaterome concept in brain health.
Collapse
Affiliation(s)
- David S Liebeskind
- Neurovascular Imaging Research Core and UCLA Stroke Center, Los Angeles, CA, USA
| |
Collapse
|
49
|
Geiseler SJ, Morland C. The Janus Face of VEGF in Stroke. Int J Mol Sci 2018; 19:ijms19051362. [PMID: 29734653 PMCID: PMC5983623 DOI: 10.3390/ijms19051362] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 04/27/2018] [Accepted: 05/01/2018] [Indexed: 12/12/2022] Open
Abstract
The family of vascular endothelial growth factors (VEGFs) are known for their regulation of vascularization. In the brain, VEGFs are important regulators of angiogenesis, neuroprotection and neurogenesis. Dysregulation of VEGFs is involved in a large number of neurodegenerative diseases and acute neurological insults, including stroke. Stroke is the main cause of acquired disabilities, and normally results from an occlusion of a cerebral artery or a hemorrhage, both leading to focal ischemia. Neurons in the ischemic core rapidly undergo necrosis. Cells in the penumbra are exposed to ischemia, but may be rescued if adequate perfusion is restored in time. The neuroprotective and angiogenic effects of VEGFs would theoretically make VEGFs ideal candidates for drug therapy in stroke. However, contradictory to what one might expect, endogenously upregulated levels of VEGF as well as the administration of exogenous VEGF is detrimental in acute stroke. This is probably due to VEGF-mediated blood–brain-barrier breakdown and vascular leakage, leading to edema and increased intracranial pressure as well as neuroinflammation. The key to understanding this Janus face of VEGF function in stroke may lie in the timing; the harmful effect of VEGFs on vessel integrity is transient, as both VEGF preconditioning and increased VEGF after the acute phase has a neuroprotective effect. The present review discusses the multifaceted action of VEGFs in stroke prevention and therapy.
Collapse
Affiliation(s)
- Samuel J Geiseler
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, 0371 Oslo, Norway.
| | - Cecilie Morland
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, 0371 Oslo, Norway.
- Institute for Behavioral Sciences, Faculty of Health Sciences, OsloMet-Oslo Metropolitan University, 0166 Oslo, Norway.
| |
Collapse
|
50
|
Xiong XY, Liu L, Yang QW. Refocusing Neuroprotection in Cerebral Reperfusion Era: New Challenges and Strategies. Front Neurol 2018; 9:249. [PMID: 29740385 PMCID: PMC5926527 DOI: 10.3389/fneur.2018.00249] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 03/28/2018] [Indexed: 12/27/2022] Open
Abstract
Pathophysiological processes of stroke have revealed that the damaged brain should be considered as an integral structure to be protected. However, promising neuroprotective drugs have failed when translated to clinical trials. In this review, we evaluated previous studies of neuroprotection and found that unsound patient selection and evaluation methods, single-target treatments, etc., without cerebral revascularization may be major reasons of failed neuroprotective strategies. Fortunately, this may be reversed by recent advances that provide increased revascularization with increased availability of endovascular procedures. However, the current improved effects of endovascular therapy are not able to match to the higher rate of revascularization, which may be ascribed to cerebral ischemia/reperfusion injury and lacking of neuroprotection. Accordingly, we suggest various research strategies to improve the lower therapeutic efficacy for ischemic stroke treatment: (1) multitarget neuroprotectant combinative therapy (cocktail therapy) should be investigated and performed based on revascularization; (2) and more efforts should be dedicated to shifting research emphasis to establish recirculation, increasing functional collateral circulation and elucidating brain–blood barrier damage mechanisms to reduce hemorrhagic transformation. Therefore, we propose that a comprehensive neuroprotective strategy before and after the endovascular treatment may speed progress toward improving neuroprotection after stroke to protect against brain injury.
Collapse
Affiliation(s)
- Xiao-Yi Xiong
- Department of Neurology, Xinqiao Hospital, The Army Medical University (Third Military Medical University), Chongqing, China
| | - Liang Liu
- Department of Neurology, Xinqiao Hospital, The Army Medical University (Third Military Medical University), Chongqing, China
| | - Qing-Wu Yang
- Department of Neurology, Xinqiao Hospital, The Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|