1
|
Zhang Y, Wang J, Zheng Z, Song S, Gu X, Yu X. Morphometrics of polypoidal choroidal vasculopathy lesions and choroidal vascular associated with treatment response using swept-source optical coherence tomography angiography. Microvasc Res 2024; 157:104759. [PMID: 39505235 DOI: 10.1016/j.mvr.2024.104759] [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: 08/16/2024] [Revised: 10/19/2024] [Accepted: 11/02/2024] [Indexed: 11/08/2024]
Abstract
PURPOSE To evaluate quantitative metrics of neovascularization lesions and choroidal vascular using swept-source optical coherence tomography angiography (SS-OCTA) in polypoidal choroidal vasculopathy (PCV) eyes, and investigate the relationship between imaging biomarkers and treatment outcomes of intravitreal anti-vascular endothelial growth factor (VEGF). METHODS We retrospectively recruited 56 PCV patients. Choroidal features included subfoveal choroidal thickness (SFCT) and choroidal vascularity index (CVI). Quantitative metrics of neovascularization lesions included total vessel length (TVL), average vessel length (AVL), junction density (JD), total number of endpoints (TNE), and mean lacunarity (ML). We performed multivariate logistic and linear regression models to determine the prognostic factors for functional and morphological outcomes. RESULTS By comparison, functional good-responders had poorer best corrected visual acuity, higher TNE, and lower ML at baseline. Morphological good-responders had higher central retinal thickness, higher TNE, lower TVL and AVL, lower ML, lower SFCT and CVI. High-shrinkage of vessel area subgroup had higher JD and TNE, lower TVL and AVL, lower ML, lower SFCT and CVI. Multivariate analysis showed good morphological response was correlated with lower SFCT (P < 0.01). High-shrinkage subgroup was correlated with lower AVL (P = 0.017) and higher TNE (P < 0.01). CONCLUSION Quantitative metrics of neovascularization lesions and choroidal characteristics using SS-OCTA had the potential to be imaging biomarkers for predicting the response to anti-VEGF treatment. PCV lesions with higher TNE and lower AVL tended to appear higher shrinkage of vessel area, and lower SFCT was correlated with good morphological response.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Ophthalmology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China; Graduate School of Peking Union Medical College, Beijing, China
| | - Jianing Wang
- Department of Ophthalmology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China; Graduate School of Peking Union Medical College, Beijing, China
| | - Zhaoxia Zheng
- Department of Ophthalmology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China; Graduate School of Peking Union Medical College, Beijing, China
| | - Shuang Song
- Department of Ophthalmology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaoya Gu
- Department of Ophthalmology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaobing Yu
- Department of Ophthalmology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China; Graduate School of Peking Union Medical College, Beijing, China.
| |
Collapse
|
2
|
Ghanbari M, Salkovskiy Y, Carlson MA. The rat as an animal model in chronic wound research: An update. Life Sci 2024; 351:122783. [PMID: 38848945 DOI: 10.1016/j.lfs.2024.122783] [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: 01/10/2024] [Revised: 03/29/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024]
Abstract
The increasing global prevalence of chronic wounds underscores the growing importance of developing effective animal models for their study. This review offers a critical evaluation of the strengths and limitations of rat models frequently employed in chronic wound research and proposes potential improvements. It explores these models in the context of key comorbidities, including diabetes, venous and arterial insufficiency, pressure-induced blood flow obstruction, and infections. Additionally, the review examines important wound factors including age, sex, smoking, and the impact of anesthetic and analgesic drugs, acknowledging their substantial effects on research outcomes. A thorough understanding of these variables is crucial for refining animal models and can provide valuable insights for future research endeavors.
Collapse
Affiliation(s)
- Mahboubeh Ghanbari
- Department of Biomechanics, University of Nebraska at Omaha, Omaha, NE, USA.
| | - Yury Salkovskiy
- Department of Biomechanics, University of Nebraska at Omaha, Omaha, NE, USA.
| | - Mark A Carlson
- Department of Surgery, Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
3
|
Memarian P, Bagher Z, Asghari S, Aleemardani M, Seifalian A. Emergence of graphene as a novel nanomaterial for cardiovascular applications. NANOSCALE 2024; 16:12793-12819. [PMID: 38919053 DOI: 10.1039/d4nr00018h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/27/2024]
Abstract
Cardiovascular diseases (CDs) are the foremost cause of death worldwide. Several promising therapeutic methods have been developed for this approach, including pharmacological, surgical intervention, cell therapy, or biomaterial implantation since heart tissue is incapable of regenerating and healing on its own. The best treatment for heart failure to date is heart transplantation and invasive surgical intervention, despite their invasiveness, donor limitations, and the possibility of being rejected by the patient's immune system. To address these challenges, research is being conducted on less invasive and efficient methods. Consequently, graphene-based materials (GBMs) have attracted a great deal of interest in the last decade because of their exceptional mechanical, electrical, chemical, antibacterial, and biocompatibility properties. An overview of GBMs' applications in the cardiovascular system has been presented in this article. Following a brief explanation of graphene and its derivatives' properties, the potential of GBMs to improve and restore cardiovascular system function by using them as cardiac tissue engineering, stents, vascular bypass grafts,and heart valve has been discussed.
Collapse
Affiliation(s)
- Paniz Memarian
- Nanotechnology and Regenerative Medicine Commercialization Centre, London BioScience Innovation Centre, London, UK.
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Zohreh Bagher
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Department of Tissue Engineering & Regenerative Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sheida Asghari
- Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| | - Mina Aleemardani
- Biomaterials and Tissue Engineering Group, Department of Materials Science and Engineering, Kroto Research Institute, The University of Sheffield, Sheffield, S3 7HQ, UK.
- Department of Translational Health Science, Bristol Medical School, University of Bristol, Bristol BS1 3NY, UK.
| | - Alexander Seifalian
- Nanotechnology and Regenerative Medicine Commercialization Centre, London BioScience Innovation Centre, London, UK.
| |
Collapse
|
4
|
Kukharchuk O, Bopardikar A, Anand Baskaran PP, Kukharchuk A, Kulkarni R, Ranbhor R. Fetal progenitor cells for treatment of chronic limb ischemia. AMERICAN JOURNAL OF STEM CELLS 2024; 13:169-190. [PMID: 39021376 PMCID: PMC11249671 DOI: 10.62347/mzki8393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 05/06/2024] [Indexed: 07/20/2024]
Abstract
OBJECTIVES This study investigated the therapeutic potential of fetal progenitor cells (FPCs) in the treatment of chronic non-healing wounds and ulcers associated with chronic limb ischemia (CLI). The research aimed to elucidate the mechanism of action of FPCs and evaluate their efficacy and safety in CLI patients. METHODS The researchers isolated FPCs from aborted human fetal liver, brain, and skin tissues and thoroughly characterized them. The preclinical phase of the study involved assessing the effects of FPCs in a rat model of CLI. Subsequently, a randomized controlled clinical trial was conducted to compare the efficacy of FPCs with standard treatment and autologous bone marrow mononuclear cells in CLI patients. The clinical trial lasted 12 months, with a follow-up period of 24-36 months. The primary outcomes included wound healing, frequency of major and minor amputations, pain reduction, and the incidence of complications. Secondary outcomes involved changes in local hemodynamics and histological, ultrastructural, and immunohistochemical assessments of angiogenesis. RESULTS In the animal model, FPC treatment significantly enhanced angiogenesis and accelerated healing of ischemic wounds compared to controls. The clinical trial in CLI patients demonstrated that the FPC therapy achieved substantially higher rates of complete wound closure, prevention of major amputation, pain reduction, and improvement in ankle-brachial index compared to control groups. Notably, the study reported no serious adverse events. CONCLUSIONS FPC therapy exhibited remarkable efficacy in promoting the healing of ischemic wounds, preventing amputation, and improving symptoms and quality of life in patients with CLI. The proangiogenic and provasculogenic effects of FPCs may be attributed to their ability to secrete specific growth factors. These findings provide new insights into the development of cellular therapeutic angiogenesis as a promising approach for the treatment of peripheral arterial diseases.
Collapse
Affiliation(s)
- Oleksandr Kukharchuk
- ReeLabs Pvt. Ltd.1st Floor, KK Chambers, Sir P.T. Rd., Azad Maidan, Fort, Mumbai 400001, India
| | | | | | - Andrii Kukharchuk
- ReeLabs Pvt. Ltd.1st Floor, KK Chambers, Sir P.T. Rd., Azad Maidan, Fort, Mumbai 400001, India
| | - Rohit Kulkarni
- ReeLabs Pvt. Ltd.1st Floor, KK Chambers, Sir P.T. Rd., Azad Maidan, Fort, Mumbai 400001, India
| | | |
Collapse
|
5
|
Fu Y, Zhang Z, Webster KA, Paulus YM. Treatment Strategies for Anti-VEGF Resistance in Neovascular Age-Related Macular Degeneration by Targeting Arteriolar Choroidal Neovascularization. Biomolecules 2024; 14:252. [PMID: 38540673 PMCID: PMC10968528 DOI: 10.3390/biom14030252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/14/2024] [Accepted: 02/16/2024] [Indexed: 05/04/2024] Open
Abstract
Despite extensive use of intravitreal anti-vascular endothelial growth factor (anti-VEGF) biologics for over a decade, neovascular age-related macular degeneration (nAMD) or choroidal neovascularization (CNV) continues to be a major cause of irreversible vision loss in developed countries. Many nAMD patients demonstrate persistent disease activity or experience declining responses over time despite anti-VEGF treatment. The underlying mechanisms of anti-VEGF resistance are poorly understood, and no effective treatment strategies are available to date. Here we review evidence from animal models and clinical studies that supports the roles of neovascular remodeling and arteriolar CNV formation in anti-VEGF resistance. Cholesterol dysregulation, inflammation, and ensuing macrophage activation are critically involved in arteriolar CNV formation and anti-VEGF resistance. Combination therapy by neutralizing VEGF and enhancing cholesterol removal from macrophages is a promising strategy to combat anti-VEGF resistance in CNV.
Collapse
Affiliation(s)
- Yingbin Fu
- Cullen Eye Institute, Baylor College of Medicine, Houston, TX 77030, USA; (Z.Z.); (K.A.W.)
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Zhao Zhang
- Cullen Eye Institute, Baylor College of Medicine, Houston, TX 77030, USA; (Z.Z.); (K.A.W.)
| | - Keith A. Webster
- Cullen Eye Institute, Baylor College of Medicine, Houston, TX 77030, USA; (Z.Z.); (K.A.W.)
- Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Yannis M. Paulus
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA;
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA
| |
Collapse
|
6
|
Yang Y, Guo J, Li M, Chu G, Jin H, Ma J, Jia Q. Cancer stem cells and angiogenesis. Pathol Res Pract 2024; 253:155064. [PMID: 38160481 DOI: 10.1016/j.prp.2023.155064] [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: 12/10/2023] [Revised: 12/23/2023] [Accepted: 12/24/2023] [Indexed: 01/03/2024]
Abstract
Cancer remains the primary cause of mortality in developed nations. Although localized tumors can be effectively addressed through surgery, radiotherapy, and other targeted methods, drug efficacy often wanes in the context of metastatic diseases. As a result, significant efforts are being made to develop drugs capable of not only inhibiting tumor growth but also impeding the metastasis of malignant tumors, with a focus on hindering their migration to adjacent organs. Cancer stem cells metastasize via blood and lymphatic vessels, exhibiting a high mutation rate, significant variability, and a predisposition to drug resistance. In contrast, endothelial cells, being less prone to mutation, are less likely to give rise to drug-resistant clones. Furthermore, the direct contact of circulating anti-angiogenic drugs with vascular endothelial cells expedites their therapeutic impact. Hence, anti-angiogenesis targeted therapy assumes a pivotal role in cancer treatment. This paper provides a succinct overview of the molecular mechanisms governing the interaction between cancer stem cells and angiogenesis.
Collapse
Affiliation(s)
- Yanru Yang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Pathology, School of Basic Medicine and Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jingyu Guo
- Department of Anesthesiology, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Mingyang Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Pathology, School of Basic Medicine and Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Guangxin Chu
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Hai Jin
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China.
| | - Jing Ma
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Pathology, School of Basic Medicine and Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| | - Qingge Jia
- Department of Reproductive Medicine, Xi'an International Medical Center Hospital, Northwest University, Xi'an, China.
| |
Collapse
|
7
|
Mankuzhy P, Dharmarajan A, Perumalsamy LR, Sharun K, Samji P, Dilley RJ. The role of Wnt signaling in mesenchymal stromal cell-driven angiogenesis. Tissue Cell 2023; 85:102240. [PMID: 37879288 DOI: 10.1016/j.tice.2023.102240] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 09/28/2023] [Accepted: 10/11/2023] [Indexed: 10/27/2023]
Abstract
Development, growth, and remodeling of blood vessels occur through an intricate process involving cell differentiation, proliferation, and rearrangement by cell migration under the direction of various signaling pathways. Recent reports highlight that resident and exogenous mesenchymal stromal cells (MSCs) have the potential to regulate the neovascularization process through paracrine secretion of proangiogenic factors. Recent research has established that the vasculogenic potential of MSCs is regulated by several signaling pathways, including the Wnt signaling pathway, and their interplay. These findings emphasize the complex nature of the vasculogenic process and underscore the importance of understanding the underlying molecular mechanisms for the development of effective cell-based therapies in regenerative medicine. This review provides an updated briefing on the canonical and non-canonical Wnt signaling pathways and summarizes the recent reports of both in vitro and in vivo studies with the involvement of MSCs of various sources in the vasculogenic process mediated by Wnt signaling pathways. Here we outline the current understanding of the plausible role of the Wnt signaling pathway, specifically in MSC-regulated angiogenesis.
Collapse
Affiliation(s)
- Pratheesh Mankuzhy
- Department of Surgery and Centre for Medical Research, Faculty of Health and Medical Sciences, The University of Western Australia, 6009 Perth, Australia; College of Veterinary and Animal Sciences - Mannuthy, Kerala Veterinary and Animal Sciences University, Pookode, Wayanad, Kerala 673576 India.
| | - Arun Dharmarajan
- Department of Biomedical Sciences, Sri Ramachandra faculty of Biomedical Sciences, Technology and Research, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai 600116, India; School of Pharmacy and Biomedical Sciences, Curtin University, Bentley, Perth, Western Australia, Australia; School of Human Sciences, Faculty of Life Sciences, University of Western Australia, 6009 Perth, Australia
| | - Lakshmi R Perumalsamy
- Department of Biomedical Sciences, Sri Ramachandra faculty of Biomedical Sciences, Technology and Research, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai 600116, India
| | - Khan Sharun
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Priyanka Samji
- Department of Biomedical Sciences, Sri Ramachandra faculty of Biomedical Sciences, Technology and Research, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai 600116, India
| | - Rodney J Dilley
- Department of Surgery and Centre for Medical Research, Faculty of Health and Medical Sciences, The University of Western Australia, 6009 Perth, Australia
| |
Collapse
|
8
|
Herajärvi J, Juvonen T. Preparing the spinal cord - priming or preconditioning? A systematic review of experimental studies. Scand Cardiovasc J Suppl 2023; 57:2166100. [PMID: 36660818 DOI: 10.1080/14017431.2023.2166100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Objectives. Paraplegia is devastating complication associated with thoracic and thoracoabdominal aortic aneurysm repair. Vast evidence has been gathered on pre-, peri- and postoperative protective adjuncts aiming to minimize spinal cord ischemia. This review focuses on the pretreatment phase of open surgical or endovascular aortic procedures and gathers the experimental data on the interventional preconditioning and priming methods that increase the spinal cord ischemic tolerance. Design. By the start of March 2021, a systematic review was performed in PubMed, Scopus and Web of Science core collection to identify the articles that reported (i) either an ischemic preconditioning, remote ischemic preconditioning or priming method prior to (ii) experimental spinal cord ischemia performed in endovascular or open surgical fashion mimicking either thoracic, abdominal or thoracoabdominal aortic aneurysm procedures. (iii) The outcomes were reported via neurological, motor-evoked potential, somatosensory-evoked potential, histopathological, immunohistochemical, physiological analysis, or in different combinations of these measurements. Results. The search yielded 7802 articles, and 57 articles were included in the systematic review. The articles were assessed by the evaluated species, the utilized pretreatment, the measured protective effects, and the suggested underlying mechanisms. Conclusions. The reviewed articles showed several possible mechanisms in ischemic and remote ischemic preconditioning for prevention of spinal cord ischemia. The main suggested method for priming was arteriogenetic stimulus. Future studies should confirm these hints of arteriogenetic stimulus with more precise quantification of the protective recruitment process.
Collapse
Affiliation(s)
- Johanna Herajärvi
- Heart and Lung Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland.,Research Unit of Surgery, Anesthesia and Critical Care, University of Oulu, Oulu, Finland
| | - Tatu Juvonen
- Heart and Lung Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland.,Research Unit of Surgery, Anesthesia and Critical Care, University of Oulu, Oulu, Finland
| |
Collapse
|
9
|
Son WH, Park HT, Jeon BH, Ha MS. Moderate intensity walking exercises reduce the body mass index and vascular inflammatory factors in postmenopausal women with obesity: a randomized controlled trial. Sci Rep 2023; 13:20172. [PMID: 37978254 PMCID: PMC10656478 DOI: 10.1038/s41598-023-47403-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 11/13/2023] [Indexed: 11/19/2023] Open
Abstract
Postmenopause, the secretion of female hormones changes, causing excessive fat accumulation in the body and leading to chronic inflammation, which increases the incidence of cardiovascular diseases (CVD). Walking is an easily accessible daily exercise and effective non-pharmacological treatment for reducing obesity and the incidence of CVD. The aim of this study was to investigate the effect of moderate intensity walking exercises on body composition, vascular inflammatory factors, and vascular endothelial growth factor (VEGF) in postmenopausal women with obesity. Twenty-six older postmenopausal women with obesity (ages 68-72) were randomly assigned to control (n = 12, BMI 26.06 ± 1.37) or exercise (n = 14, BMI 26.04 ± 1.94) groups. Following a 12-week moderate intensity walking exercise program, we measured the participants' body composition with an InBody S10 analyzer and assessed blood sera using enzyme-linked immunosorbent assays. There was a significant clustering by weight (p < 0.01), body mass index (p < 0.01), percentage body fat (p < 0.001), high-sensitivity C-reactive protein (p < 0.05), interleukin-6, and tumor necrosis factor-α (p < 0.05) being significantly decreased in the exercise group. Although VEGF levels did not change significantly, a tendency to increase was observed in participants that exercised. Our results indicate that walking exercise may help prevent CVD in postmenopausal women with obesity by reducing obesity and vascular inflammatory factors.
Collapse
Affiliation(s)
- Woo-Hyeon Son
- Institute of Convergence Bio-Health, Dong-A University, 26 Daesingongwon-ro, Seo-gu, Busan, 49201, Republic of Korea
| | - Hyun-Tae Park
- Graduate School of Health Care and Sciences, College of Health Science, Dong-A University, 37, Nakdong-daero 550beon-gil, Saha-gu, Busan, 49315, Republic of Korea.
| | - Byeong Hwan Jeon
- Department of Sports and Health Science, College of Arts, Kyungsung University, 309, Suyeong-ro, Nam-gu, Busan, 48434, Republic of Korea
| | - Min-Seong Ha
- Laboratory of Sports Conditioning: Nutrition Biochemistry and Neuroscience, Department of Sports Science, College of Arts and Sports, University of Seoul, 163 Seoulsiripdaero, Dongdaemun-gu, Seoul, 02504, Republic of Korea.
| |
Collapse
|
10
|
Mohamad Yusoff F, Higashi Y. Mesenchymal Stem/Stromal Cells for Therapeutic Angiogenesis. Cells 2023; 12:2162. [PMID: 37681894 PMCID: PMC10486439 DOI: 10.3390/cells12172162] [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: 07/13/2023] [Revised: 08/18/2023] [Accepted: 08/25/2023] [Indexed: 09/09/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are known to possess medicinal properties to facilitate vascular regeneration. Recent advances in the understanding of the utilities of MSCs in physiological/pathological tissue repair and technologies in isolation, expansion, and enhancement strategies have led to the use of MSCs for vascular disease-related treatments. Various conditions, including chronic arterial occlusive disease, diabetic ulcers, and chronic wounds, cause significant morbidity in patients. Therapeutic angiogenesis by cell therapy has led to the possibilities of treatment options in promoting angiogenesis, treating chronic wounds, and improving amputation-free survival. Current perspectives on the options for the use of MSCs for therapeutic angiogenesis in vascular research and in medicine, either as a monotherapy or in combination with conventional interventions, for treating patients with peripheral artery diseases are discussed in this review.
Collapse
Affiliation(s)
- Farina Mohamad Yusoff
- Department of Regenerative Medicine, Division of Radiation Medical Science, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima 734-8553, Japan;
| | - Yukihito Higashi
- Department of Regenerative Medicine, Division of Radiation Medical Science, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima 734-8553, Japan;
- Division of Regeneration and Medicine, Hiroshima University Hospital, Hiroshima 734-8551, Japan
| |
Collapse
|
11
|
Poledniczek M, Neumayer C, Kopp CW, Schlager O, Gremmel T, Jozkowicz A, Gschwandtner ME, Koppensteiner R, Wadowski PP. Micro- and Macrovascular Effects of Inflammation in Peripheral Artery Disease-Pathophysiology and Translational Therapeutic Approaches. Biomedicines 2023; 11:2284. [PMID: 37626780 PMCID: PMC10452462 DOI: 10.3390/biomedicines11082284] [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: 06/25/2023] [Revised: 08/10/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Inflammation has a critical role in the development and progression of atherosclerosis. On the molecular level, inflammatory pathways negatively impact endothelial barrier properties and thus, tissue homeostasis. Conformational changes and destruction of the glycocalyx further promote pro-inflammatory pathways also contributing to pro-coagulability and a prothrombotic state. In addition, changes in the extracellular matrix composition lead to (peri-)vascular remodelling and alterations of the vessel wall, e.g., aneurysm formation. Moreover, progressive fibrosis leads to reduced tissue perfusion due to loss of functional capillaries. The present review aims at discussing the molecular and clinical effects of inflammatory processes on the micro- and macrovasculature with a focus on peripheral artery disease.
Collapse
Affiliation(s)
- Michael Poledniczek
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (M.P.); (C.W.K.); (O.S.); (M.E.G.); (R.K.)
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
| | - Christoph Neumayer
- Division of Vascular Surgery, Department of General Surgery, Medical University of Vienna, 1090 Vienna, Austria;
| | - Christoph W. Kopp
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (M.P.); (C.W.K.); (O.S.); (M.E.G.); (R.K.)
| | - Oliver Schlager
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (M.P.); (C.W.K.); (O.S.); (M.E.G.); (R.K.)
| | - Thomas Gremmel
- Department of Internal Medicine I, Cardiology and Intensive Care Medicine, Landesklinikum Mistelbach-Gänserndorf, 2130 Mistelbach, Austria;
- Institute of Cardiovascular Pharmacotherapy and Interventional Cardiology, Karl Landsteiner Society, 3100 St. Pölten, Austria
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biophysics, Biochemistry and Biotechnology, Jagiellonian University, 31-007 Krakow, Poland;
| | - Michael E. Gschwandtner
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (M.P.); (C.W.K.); (O.S.); (M.E.G.); (R.K.)
| | - Renate Koppensteiner
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (M.P.); (C.W.K.); (O.S.); (M.E.G.); (R.K.)
| | - Patricia P. Wadowski
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (M.P.); (C.W.K.); (O.S.); (M.E.G.); (R.K.)
| |
Collapse
|
12
|
Di X, Gao X, Peng L, Ai J, Jin X, Qi S, Li H, Wang K, Luo D. Cellular mechanotransduction in health and diseases: from molecular mechanism to therapeutic targets. Signal Transduct Target Ther 2023; 8:282. [PMID: 37518181 PMCID: PMC10387486 DOI: 10.1038/s41392-023-01501-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 08/01/2023] Open
Abstract
Cellular mechanotransduction, a critical regulator of numerous biological processes, is the conversion from mechanical signals to biochemical signals regarding cell activities and metabolism. Typical mechanical cues in organisms include hydrostatic pressure, fluid shear stress, tensile force, extracellular matrix stiffness or tissue elasticity, and extracellular fluid viscosity. Mechanotransduction has been expected to trigger multiple biological processes, such as embryonic development, tissue repair and regeneration. However, prolonged excessive mechanical stimulation can result in pathological processes, such as multi-organ fibrosis, tumorigenesis, and cancer immunotherapy resistance. Although the associations between mechanical cues and normal tissue homeostasis or diseases have been identified, the regulatory mechanisms among different mechanical cues are not yet comprehensively illustrated, and no effective therapies are currently available targeting mechanical cue-related signaling. This review systematically summarizes the characteristics and regulatory mechanisms of typical mechanical cues in normal conditions and diseases with the updated evidence. The key effectors responding to mechanical stimulations are listed, such as Piezo channels, integrins, Yes-associated protein (YAP) /transcriptional coactivator with PDZ-binding motif (TAZ), and transient receptor potential vanilloid 4 (TRPV4). We also reviewed the key signaling pathways, therapeutic targets and cutting-edge clinical applications of diseases related to mechanical cues.
Collapse
Affiliation(s)
- Xingpeng Di
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Xiaoshuai Gao
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Liao Peng
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Jianzhong Ai
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Xi Jin
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Shiqian Qi
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Hong Li
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Kunjie Wang
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China.
| | - Deyi Luo
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China.
| |
Collapse
|
13
|
Lamin V, Mani AM, Singh MV, Dokun AO. Endothelial Progenitor Cells and Macrophage Subsets Recruitment in Postischemic Mouse Hind Limbs. J Vasc Res 2023; 60:148-159. [PMID: 37336198 DOI: 10.1159/000530732] [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: 07/01/2022] [Accepted: 03/14/2023] [Indexed: 06/21/2023] Open
Abstract
INTRODUCTION Peripheral arterial disease (PAD) occurs from atherosclerotic obstruction of arteries in the lower extremities. Restoration of perfusion requires angiogenesis and arteriogenesis through migration and differentiation of endothelial progenitor cells (EPCs) and macrophages at the site of injury. The time of recruitment has not been fully investigated. In this study, we investigated the infiltration of these cells in murine hind limb ischemia (HLI) model of PAD. METHODS EPCs and M1-like and M2-like macrophages from ischemic skeletal muscles were quantified by flow cytometry at day-0, 1, 3, 7, and 14 post-HLI. RESULTS The abundance of EPCs increased from day 1 and was highest on day 7 until day 14. M1-like population similarly increased and was highest on day 14 during the experiment. M2-like population was significantly greater than M1-like at baseline but surpassed the highest value of M1-like by day 7 during the experiment. Muscle regeneration and capillary density also increased and were highest at days 3 and 7, respectively, during the experiment. All mice achieved near full perfusion recovery by day 14. CONCLUSION Thus, we observed a gradual increase in the percentage of EPC's and this was temporally paralleled with initial increase in M1-like followed by sustained increased in M2-like macrophages and perfusion recovered post-HLI.
Collapse
Affiliation(s)
- Victor Lamin
- Division of Endocrinology and Metabolism, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Arul M Mani
- Division of Endocrinology and Metabolism, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Madhu V Singh
- Division of Endocrinology and Metabolism, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Ayotunde O Dokun
- Division of Endocrinology and Metabolism, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
14
|
Yoo H, La H, Park C, Yoo S, Lee H, Song H, Do JT, Choi Y, Hong K. Common and distinct functions of mouse Dot1l in the regulation of endothelial transcriptome. Front Cell Dev Biol 2023; 11:1176115. [PMID: 37397258 PMCID: PMC10311421 DOI: 10.3389/fcell.2023.1176115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 06/06/2023] [Indexed: 07/04/2023] Open
Abstract
Epigenetic mechanisms are mandatory for endothelial called lymphangioblasts during cardiovascular development. Dot1l-mediated gene transcription in mice is essential for the development and function of lymphatic ECs (LECs). The role of Dot1l in the development and function of blood ECs blood endothelial cells is unclear. RNA-seq datasets from Dot1l-depleted or -overexpressing BECs and LECs were used to comprehensively analyze regulatory networks of gene transcription and pathways. Dot1l depletion in BECs changed the expression of genes involved in cell-to-cell adhesion and immunity-related biological processes. Dot1l overexpression modified the expression of genes involved in different types of cell-to-cell adhesion and angiogenesis-related biological processes. Genes involved in specific tissue development-related biological pathways were altered in Dot1l-depleted BECs and LECs. Dot1l overexpression altered ion transportation-related genes in BECs and immune response regulation-related genes in LECs. Importantly, Dot1l overexpression in BECs led to the expression of genes related to the angiogenesis and increased expression of MAPK signaling pathways related was found in both Dot1l-overexpressing BECs and LECs. Therefore, our integrated analyses of transcriptomics in Dot1l-depleted and Dot1l-overexpressed ECs demonstrate the unique transcriptomic program of ECs and the differential functions of Dot1l in the regulation of gene transcription in BECs and LECs.
Collapse
|
15
|
Özsoyler İ, Uçak HA, Badak TO, Çakallıoğlu A, Bayraktar M, Arslan AS. The impact of the apelinergic system in coronary collateral formation. TURK GOGUS KALP DAMAR CERRAHISI DERGISI 2023; 31:192-198. [PMID: 37484641 PMCID: PMC10357849 DOI: 10.5606/tgkdc.dergisi.2023.24422] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 03/23/2023] [Indexed: 07/25/2023]
Abstract
Background This study aims to examine the relationship between the development of coronary collateral circulation and serum elabela levels. Methods Between January 2020 and December 2021, a total of 50 control individuals (29 males, 21 females; mean age: 63.2±10.0 years; range, 52 to 73 years) with no significant coronary artery disease as confirmed by angiography (Group 1) and 100 patients (55 males, 45 females; mean age: 66.6±9.6 years; range, 56 to 75 years) with coronary artery disease were included. The patients were further divided into two equal groups according to the Rentrop classification as poor (Group 2) and good coronary collateral circulation (Group 3). All groups were compared in terms of several parameters, particularly serum elabela levels. Results Serum elabela levels were found to be statistically higher in the group with good collateral than the other groups (p<0.05). Low serum elabela levels increased the risk of developing weak collaterals by 2.43 times. Conclusion The elabela protein is directly related to good collateral development and can be considered a potential agent for treatment.
Collapse
Affiliation(s)
- İbrahim Özsoyler
- Department of Cardiovascular Surgery, Health Sciences University, Adana Şehir Training and Research Hospital, Adana, Türkiye
| | - Haci Ali Uçak
- Department of Cardiovascular Surgery, Health Sciences University, Adana Şehir Training and Research Hospital, Adana, Türkiye
| | - Tolga Onur Badak
- Department of Cardiovascular Surgery, Health Sciences University, Adana Şehir Training and Research Hospital, Adana, Türkiye
| | - Ahmet Çakallıoğlu
- Department of Cardiovascular Surgery, Health Sciences University, Adana Şehir Training and Research Hospital, Adana, Türkiye
| | - Muhammet Bayraktar
- Department of Public Health, Niğde Ömer Halisdemir University Faculty of Medicine, Niğde, Türkiye
| | - Ahmet Süha Arslan
- Department of Cardiovascular Surgery, Health Sciences University, Adana Şehir Training and Research Hospital, Adana, Türkiye
| |
Collapse
|
16
|
Pascale JV, Wolf A, Kadish Y, Diegisser D, Kulaprathazhe MM, Yemane D, Ali S, Kim N, Baruch DE, Yahaya MAF, Dirice E, Adebesin AM, Falck JR, Schwartzman ML, Garcia V. 20-Hydroxyeicosatetraenoic acid (20-HETE): Bioactions, receptors, vascular function, cardiometabolic disease and beyond. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2023; 97:229-255. [PMID: 37236760 PMCID: PMC10683332 DOI: 10.1016/bs.apha.2023.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2023]
Abstract
Vascular function is dynamically regulated and dependent on a bevy of cell types and factors that work in concert across the vasculature. The vasoactive eicosanoid, 20-Hydroxyeicosatetraenoic acid (20-HETE) is a key player in this system influencing the sensitivity of the vasculature to constrictor stimuli, regulating endothelial function, and influencing the renin angiotensin system (RAS), as well as being a driver of vascular remodeling independent of blood pressure elevations. Several of these bioactions are accomplished through the ligand-receptor pairing between 20-HETE and its high-affinity receptor, GPR75. This 20-HETE axis is at the root of various vascular pathologies and processes including ischemia induced angiogenesis, arteriogenesis, septic shock, hypertension, atherosclerosis, myocardial infarction and cardiometabolic diseases including diabetes and insulin resistance. Pharmacologically, several preclinical tools have been developed to disrupt the 20-HETE axis including 20-HETE synthesis inhibitors (DDMS and HET0016), synthetic 20-HETE agonist analogues (20-5,14-HEDE and 20-5,14-HEDGE) and 20-HETE receptor blockers (AAA and 20-SOLA). Systemic or cell-specific therapeutic targeting of the 20-HETE-GPR75 axis continues to be an invaluable approach as studies examine the molecular underpinnings activated by 20-HETE under various physiological settings. In particular, the development and characterization of 20-HETE receptor blockers look to be a promising new class of compounds that can provide a considerable benefit to patients suffering from these cardiovascular pathologies.
Collapse
Affiliation(s)
- Jonathan V Pascale
- Department of Pharmacology, New York Medical College, Valhalla, NY, United States
| | - Alexandra Wolf
- Department of Pharmacology, New York Medical College, Valhalla, NY, United States
| | - Yonaton Kadish
- School of Medicine, New York Medical College, Valhalla, NY, United States
| | - Danielle Diegisser
- Department of Pharmacology, New York Medical College, Valhalla, NY, United States
| | | | - Danait Yemane
- Department of Pharmacology, New York Medical College, Valhalla, NY, United States
| | - Samir Ali
- School of Medicine, New York Medical College, Valhalla, NY, United States
| | - Namhee Kim
- School of Medicine, New York Medical College, Valhalla, NY, United States
| | - David E Baruch
- School of Medicine, New York Medical College, Valhalla, NY, United States
| | - Muhamad Afiq Faisal Yahaya
- Department of Basic Sciences, MAHSA University, Selangor Darul Ehsan, Malaysia; Department of Human Anatomy, Universiti Putra Malaysia (UPM), Selangor Darul Ehsan, Malaysia
| | - Ercument Dirice
- Department of Pharmacology, New York Medical College, Valhalla, NY, United States
| | - Adeniyi M Adebesin
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - John R Falck
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Michal L Schwartzman
- Department of Pharmacology, New York Medical College, Valhalla, NY, United States
| | - Victor Garcia
- Department of Pharmacology, New York Medical College, Valhalla, NY, United States.
| |
Collapse
|
17
|
Li W, Xu P, Kong L, Feng S, Shen N, Huang H, Wang W, Xu X, Wang X, Wang G, Zhang Y, Sun W, Hu W, Liu X. Elabela-APJ axis mediates angiogenesis via YAP/TAZ pathway in cerebral ischemia/reperfusion injury. Transl Res 2023; 257:78-92. [PMID: 36813109 DOI: 10.1016/j.trsl.2023.02.002] [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: 08/02/2022] [Revised: 01/13/2023] [Accepted: 02/02/2023] [Indexed: 02/24/2023]
Abstract
Angiogenesis helps to improve neurological recovery by repairing damaged brain tissue and restoring cerebral blood flow (CBF). The role of the Elabela (ELA)-Apelin receptor (APJ) system in angiogenesis has gained much attention. We aimed to investigate the function of endothelial ELA on postischemic cerebral angiogenesis. Here, we demonstrated that the endothelial ELA expression was upregulated in the ischemic brain and treatment with ELA-32 mitigated brain injury and enhanced the restoration of CBF and newly formed functional vessels following cerebral ischemia/reperfusion (I/R) injury. Furthermore, ELA-32 incubation potentiated proliferation, migration, and tube formation abilities of the mouse brain endothelial cells (bEnd.3 cells) under oxygen-glucose deprivation/reoxygenation (OGD/R) condition. RNA sequencing analysis indicated that ELA-32 incubation had a role in the Hippo signaling pathway, and improved angiogenesis-related gene expression in OGD/R-exposed bEnd.3 cells. Mechanistically, we depicted that ELA could bind to APJ and subsequently activate YAP/TAZ signaling pathway. Silence of APJ or pharmacological blockade of YAP abolished the pro-angiogenesis effects of ELA-32. Together, these findings highlight the ELA-APJ axis as a potential therapeutic strategy for ischemic stroke by showing how activation of this pathway promotes poststroke angiogenesis.
Collapse
Affiliation(s)
- Wenyu Li
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Pengfei Xu
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| | - Lingqi Kong
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Shuo Feng
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Nan Shen
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Hongmei Huang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Wuxuan Wang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiang Xu
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Xinyue Wang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Guoping Wang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Yan Zhang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Wen Sun
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Wei Hu
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Xinfeng Liu
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
18
|
Novel insights into the angiogenic function of JMJD2B in diabetic hind limb ischemia: involvement of activating Wnt/β-catenin pathway. Hum Cell 2023; 36:1011-1023. [PMID: 36773117 DOI: 10.1007/s13577-023-00874-x] [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: 10/18/2022] [Accepted: 02/01/2023] [Indexed: 02/12/2023]
Abstract
Critical limb ischemia (CLI) is a major health problem, in which diabetes is a risk factor. Lysine Demethylase 4B (JMJD2B) is a histone demethylase. Diabetic CLI model was established in mice by streptozotocin injection and femoral artery ligation. Reduced expression of JMJD2B in lower limb muscles was observed in CLI mice with or without diabetes, accompanied by impaired blood perfusion and mobility. Adenovirus-mediated JMJD2B overexpression improved blood perfusion and angiogenesis as indicated by the alternation in CD31, α-SMA, and VEGFA expression in the lower limb of diabetic mice with CLI. In vitro, JMJD2B expression and the proliferation and tube formation ability were inhibited by high glucose and ischemic conditions in HMEC-1 cells. Overexpressed-JMJD2B contributed to angiogenesis by promoting cell proliferation, migration, and tube formation of HMEC-1 cells, as well as increasing VEGFA and SDF-1 expression. Mechanism study indicated that JMJD2B overexpression activated the Wnt/β-catenin pathway by promoting β-catenin nuclear translocation and the expression. This might lead to stimulated angiogenesis, as demonstrated by the Wnt/β-catenin inhibitor XAV-939. Overall, our study revealed that JMJD2B was down-regulated in CLI mice with diabetes and JMJD2B overexpression promoted angiogenesis probably via the activation of Wnt/β-catenin pathway.
Collapse
|
19
|
Leng X, Leung TW. Collateral Flow in Intracranial Atherosclerotic Disease. Transl Stroke Res 2023; 14:38-52. [PMID: 35672561 DOI: 10.1007/s12975-022-01042-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/27/2022] [Accepted: 05/26/2022] [Indexed: 01/31/2023]
Abstract
Intracranial atherosclerotic disease (ICAD) is a major cause of ischemic stroke and transient ischemic attack (TIA) worldwide. The culprit of ICAD is frequently a high-grade intracranial atherosclerotic stenosis (ICAS) pertaining to the infarct territory, and by then, the ICAS is described as symptomatic. A high-grade ICAS may progressively limit cerebral perfusion downstream, demanding collateral compensation. Collateral circulation refers to the pre-existing and dynamic emergence of vascular channels that maintain and compensate for a failing principal vascular route. Collaterals through the Circle of Willis and leptomeningeal circulation are of utmost importance in this regard. In this article, we first discussed the epidemiology, stroke mechanisms, contemporary therapeutics, and prognosis of symptomatic ICAD. Then, we reviewed the collateral routes in ICAS, factors associated with recruitment and development of the collaterals and diagnostic imaging modalities in assessing the origin and function of collateral circulation. We discussed the associations between collateral circulation and clinical outcomes after acute reperfusion treatment in ICAD-related ischemic strokes with or without large vessel occlusion (LVO). We also conducted a systematic review and meta-analysis on the associations of collateral circulation with the risk of recurrent stroke and the functional outcome in symptomatic ICAS patients on medical treatment as secondary stroke prevention. Finally, we summarized current evidence in these aspects and proposed the future directions.
Collapse
Affiliation(s)
- Xinyi Leng
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, The Prince of Wales Hospital, Hong Kong SAR, China
| | - Thomas W Leung
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, The Prince of Wales Hospital, Hong Kong SAR, China.
| |
Collapse
|
20
|
Advances in the Physico-Chemical, Antimicrobial and Angiogenic Properties of Graphene-Oxide/Cellulose Nanocomposites for Wound Healing. Pharmaceutics 2023; 15:pharmaceutics15020338. [PMID: 36839660 PMCID: PMC9961167 DOI: 10.3390/pharmaceutics15020338] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/10/2023] [Accepted: 01/12/2023] [Indexed: 01/21/2023] Open
Abstract
Graphene oxide (GO) and its reduced form (rGO) have recently attracted a fascinating interest due to their physico-chemical properties, which have opened up new and interesting opportunities in a wide range of biomedical applications, such as wound healing. It is worth noting that GO and rGO may offer a convenient access to its ready dispersion within various polymeric matrices (such as cellulose and its derivative forms), owing to their large surface area, based on a carbon skeleton with many functional groups (i.e., hydroxyl, carboxyl, epoxy bridge, and carbonyl moieties). This results in new synergic properties due to the presence of both components (GO or rGO and polymers), acting at different length-scales. Furthermore, they have shown efficient antimicrobial and angiogenic properties, mostly related to the intracellular formation of reactive oxygen species (ROS), which are advantageous in wound care management. For this reason, GO or rGO integration in cellulose-based matrixes have allowed for designing highly advanced multifunctional hybrid nanocomposites with tailored properties. The current review aims to discuss a potential relationship between structural and physico-chemical properties (i.e., size, edge density, surface chemistry, hydrophilicity) of the nanocomposites with antimicrobials and angiogenic mechanisms that synergically influence the wound healing phenomenon, by paying particular attention to recent findings of GO or rGO/cellulose nanocomposites. Accordingly, after providing a general overview of cellulose and its derivatives, the production methods used for GO and rGO synthesis, the mechanisms that guide antimicrobial and angiogenic processes of tissue repair, as well as the most recent and remarkable outcomes on GO/cellulose scaffolds in wound healing applications, will be presented.
Collapse
|
21
|
Apeldoorn C, Safaei S, Paton J, Maso Talou GD. Computational models for generating microvascular structures: Investigations beyond medical imaging resolution. WIREs Mech Dis 2023; 15:e1579. [PMID: 35880683 PMCID: PMC10077909 DOI: 10.1002/wsbm.1579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 06/22/2022] [Accepted: 06/29/2022] [Indexed: 01/31/2023]
Abstract
Angiogenesis, arteriogenesis, and pruning are revascularization processes essential to our natural vascular development and adaptation, as well as central players in the onset and development of pathologies such as tumoral growth and stroke recovery. Computational modeling allows for repeatable experimentation and exploration of these complex biological processes. In this review, we provide an introduction to the biological understanding of the vascular adaptation processes of sprouting angiogenesis, intussusceptive angiogenesis, anastomosis, pruning, and arteriogenesis, discussing some of the more significant contributions made to the computational modeling of these processes. Each computational model represents a theoretical framework for how biology functions, and with rises in computing power and study of the problem these frameworks become more accurate and complete. We highlight physiological, pathological, and technological applications that can be benefit from the advances performed by these models, and we also identify which elements of the biology are underexplored in the current state-of-the-art computational models. This article is categorized under: Cancer > Computational Models Cardiovascular Diseases > Computational Models.
Collapse
Affiliation(s)
- Cameron Apeldoorn
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Soroush Safaei
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Julian Paton
- Cardiovascular Autonomic Research Cluster, Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Gonzalo D Maso Talou
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
22
|
Babu M, Devi D, Mäkinen P, Örd T, Aavik E, Kaikkonen M, Ylä-Herttuala S. ApoA-I Nanotherapy Rescues Postischemic Vascular Maladaptation by Modulating Endothelial Cell and Macrophage Phenotypes in Type 2 Diabetic Mice. Arterioscler Thromb Vasc Biol 2023; 43:e46-e61. [PMID: 36384268 DOI: 10.1161/atvbaha.122.318196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
BACKGROUND Diabetes is a major risk factor for peripheral arterial disease. Clinical and preclinical studies suggest an impaired collateral remodeling and angiogenesis in response to atherosclerotic arterial occlusion in diabetic conditions, although the underlying mechanisms are poorly understood. OBJECTIVE To clarify the cellular and molecular mechanisms underlying impaired postischemic adaptive vascular responses and to evaluate rHDL (reconstituted HDL)-ApoA-I nanotherapy to rescue the defect in type 2 diabetic mouse model of hindlimb ischemia. METHODS AND RESULTS Hindlimb ischemia was induced by unilateral femoral artery ligation. Collateral and capillary parameters together with blood flow recovery were analyzed from normoxic adductor and ischemic gastrocnemius muscles, respectively, at day 3 and 7 post-ligation. In response to femoral artery ligation, collateral lumen area was significantly reduced in normoxic adductor muscles. Distally, ischemic gastrocnemius muscles displayed impaired perfusion recovery and angiogenesis paralleled with persistent inflammation. Muscle-specific mRNA sequencing revealed differential expression of genes critical for smooth muscle proliferation and sprouting angiogenesis in normoxic adductor and ischemic gastrocnemius, respectively, at day 7 post-ligation. Genes typical for macrophage (Mϕ) subsets were differentially expressed across both muscle types. Cell-specific gene expression, flow cytometry, and immunohistochemistry revealed persistent IFN-I response gene upregulation in arterial endothelial cells, ECs and Mϕs from T2DM mice associated with impaired collateral remodeling, angiogenesis and perfusion recovery. Furthermore, rHDL nanotherapy rescued impaired collateral remodeling and angiogenesis through dampening EC and Mϕ inflammation in T2DM mice. CONCLUSIONS Our results suggest that an impaired collateral remodeling and sprouting angiogenesis in T2DM mice is associated with persistent IFN-I response in ECs and Mϕs. Dampening persistent inflammation and skewing ECs and Mϕ phenotype toward less inflammatory ones using rHDL nanotherapy may serve as a potential therapeutic target for T2DM peripheral arterial disease.
Collapse
Affiliation(s)
- Mohan Babu
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute, University of Eastern Finland, Kuopio (M.B., D.D., P.M., T.O., E.A., M.K., S.Y.-H.)
| | - Durga Devi
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute, University of Eastern Finland, Kuopio (M.B., D.D., P.M., T.O., E.A., M.K., S.Y.-H.)
| | - Petri Mäkinen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute, University of Eastern Finland, Kuopio (M.B., D.D., P.M., T.O., E.A., M.K., S.Y.-H.)
| | - Tiit Örd
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute, University of Eastern Finland, Kuopio (M.B., D.D., P.M., T.O., E.A., M.K., S.Y.-H.)
| | - Einari Aavik
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute, University of Eastern Finland, Kuopio (M.B., D.D., P.M., T.O., E.A., M.K., S.Y.-H.)
| | - Minna Kaikkonen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute, University of Eastern Finland, Kuopio (M.B., D.D., P.M., T.O., E.A., M.K., S.Y.-H.)
| | - Seppo Ylä-Herttuala
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute, University of Eastern Finland, Kuopio (M.B., D.D., P.M., T.O., E.A., M.K., S.Y.-H.).,Heart Center and Gene Therapy Unit, Kuopio University Hospital, Finland (S.Y.-H.)
| |
Collapse
|
23
|
Sieland J, Niederer D, Engeroff T, Vogt L, Troidl C, Schmitz-Rixen T, Banzer W, Troidl K. Changes in miRNA expression in patients with peripheral arterial vascular disease during moderate- and vigorous-intensity physical activity. Eur J Appl Physiol 2023; 123:645-654. [PMID: 36418750 PMCID: PMC9684818 DOI: 10.1007/s00421-022-05091-2] [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: 09/07/2021] [Accepted: 11/09/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Walking is the preferred therapy for peripheral arterial disease in early stage. An effect of walking exercise is the increase of blood flow and fluid shear stress, leading, triggered by arteriogenesis, to the formation of collateral blood vessels. Circulating micro-RNA may act as an important information transmitter in this process. We investigated the acute effects of a single bout of 1) aerobic walking with moderate intensity; and 2) anaerobic walking with vigorous intensity on miRNA parameters related to vascular collateral formation. METHODS Ten (10) patients with peripheral arterial disease with claudication (age 72 ± 7 years) participated in this two-armed, randomized-balanced cross-over study. The intervention arms were single bouts of supervised walking training at (1) vigorous intensity on a treadmill up to volitional exhaustion and (2) moderate intensity with individual selected speed for a duration of 20 min. One week of washout was maintained between the arms. During each intervention, heart rate was continuously monitored. Acute effects on circulating miRNAs and lactate concentration were determined using pre- and post-intervention measurement comparisons. RESULTS Vigorous-intensity walking resulted in a higher heart rate (125 ± 21 bpm) than the moderate-intensity intervention (88 ± 9 bpm) (p < 0.05). Lactate concentration was increased after vigorous-intensity walking (p = 0.005; 3.3 ± 1.2 mmol/l), but not after moderate exercising (p > 0.05; 1.7 ± 0.6 mmol/l). The circulating levels of miR-142-5p and miR-424-5p were up-regulated after moderate-intensity (p < 0.05), but not after vigorous-intensity training (p > 0.05). CONCLUSION Moderate-intensity walking seems to be more feasible than vigorous exercises to induce changes of blood flow and endurance training-related miRNAs in patients with peripheral arterial disease. Our data thus indicates that effect mechanisms might follow an optimal rather than a maximal dose response relation. Steady state walking without the necessity to reach exhaustion seems to be better suited as stimulus.
Collapse
Affiliation(s)
- Johanna Sieland
- Department of Sports Medicine, Institute of Sports Sciences, Goethe University, Ginnheimer Landstraße 39, 60487, Frankfurt, Germany.
| | - Daniel Niederer
- grid.7839.50000 0004 1936 9721Department of Sports Medicine, Institute of Sports Sciences, Goethe University, Ginnheimer Landstraße 39, 60487 Frankfurt, Germany ,grid.7839.50000 0004 1936 9721Division of Preventive and Sports Medicine, Institute for Occupational Medicine, Social Medicine and Environmental Medicine, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Tobias Engeroff
- grid.7839.50000 0004 1936 9721Institute for Occupational Medicine, Social Medicine and Environmental Medicine, Division Health and Performance, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Lutz Vogt
- grid.7839.50000 0004 1936 9721Department of Sports Medicine, Institute of Sports Sciences, Goethe University, Ginnheimer Landstraße 39, 60487 Frankfurt, Germany
| | - Christian Troidl
- grid.8664.c0000 0001 2165 8627Department of Experimental Cardiology, Medical Faculty, Justus-Liebig-University, 35392 Giessen, Germany ,grid.419757.90000 0004 0390 5331Department of Cardiology, Kerckhoff Heart and Thorax Center, 61231 Bad Nauheim, Germany ,grid.452396.f0000 0004 5937 5237German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt Am Main, Germany
| | - Thomas Schmitz-Rixen
- grid.411088.40000 0004 0578 8220Department of Vascular and Endovascular Surgery, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Winfried Banzer
- grid.7839.50000 0004 1936 9721Division of Preventive and Sports Medicine, Institute for Occupational Medicine, Social Medicine and Environmental Medicine, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Kerstin Troidl
- grid.411088.40000 0004 0578 8220Department of Vascular and Endovascular Surgery, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany ,grid.449744.e0000 0000 9323 0139Department of Life Sciences and Engineering, TH Bingen, Berlinstrasse 109, 55411 Bingen Am Rhein, Germany
| |
Collapse
|
24
|
Bui TA, Jickling GC, Winship IR. Neutrophil dynamics and inflammaging in acute ischemic stroke: A transcriptomic review. Front Aging Neurosci 2022; 14:1041333. [PMID: 36620775 PMCID: PMC9813499 DOI: 10.3389/fnagi.2022.1041333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 11/28/2022] [Indexed: 12/24/2022] Open
Abstract
Stroke is among the leading causes of death and disability worldwide. Restoring blood flow through recanalization is currently the only acute treatment for cerebral ischemia. Unfortunately, many patients that achieve a complete recanalization fail to regain functional independence. Recent studies indicate that activation of peripheral immune cells, particularly neutrophils, may contribute to microcirculatory failure and futile recanalization. Stroke primarily affects the elderly population, and mortality after endovascular therapies is associated with advanced age. Previous analyses of differential gene expression across injury status and age identify ischemic stroke as a complex age-related disease. It also suggests robust interactions between stroke injury, aging, and inflammation on a cellular and molecular level. Understanding such interactions is crucial in developing effective protective treatments. The global stroke burden will continue to increase with a rapidly aging human population. Unfortunately, the mechanisms of age-dependent vulnerability are poorly defined. In this review, we will discuss how neutrophil-specific gene expression patterns may contribute to poor treatment responses in stroke patients. We will also discuss age-related transcriptional changes that may contribute to poor clinical outcomes and greater susceptibility to cerebrovascular diseases.
Collapse
Affiliation(s)
- Truong An Bui
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Glen C. Jickling
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Department of Medicine, Division of Neurology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Ian R. Winship
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
25
|
Kwon M, Robins L, McGlynn ML, Collins C, Pekas EJ, Park SY, Slivka D. No Mitochondrial Related Transcriptional Changes in Human Skeletal Muscle after Local Heat Application. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:17051. [PMID: 36554930 PMCID: PMC9779680 DOI: 10.3390/ijerph192417051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/13/2022] [Accepted: 12/16/2022] [Indexed: 06/17/2023]
Abstract
The purpose of the study is to determine the impact of local heating on skeletal muscle transcriptional response related to mitochondrial biogenesis and mitophagy. Twelve healthy subjects (height, 176.0 ± 11.9 cm; weight, 83.6 ± 18.3 kg; and body composition, 19.0 ± 7.7% body fat) rested in a semi-reclined position for 4 h with a heated thermal wrap (HOT) around one thigh and a wrap without temperature regulation (CON) around the other (randomized). Skin temperature, blood flow, intramuscular temperature, and a skeletal muscle biopsy from the vastus lateralis were obtained after the 4 h intervention. Skin temperature via infrared thermometer and thermal camera was higher after HOT (37.3 ± 0.7 and 36.7 ± 1.0 °C, respectively) than CON (34.8 ± 0.7, 35.2 ± 0.8 °C, respectively, p < 0.001). Intramuscular temperature was higher in HOT (36.3 ± 0.4 °C) than CON (35.2 ± 0.8 °C, p < 0.001). Femoral artery blood flow was higher in HOT (304.5 ± 12.5 mL‧min-1) than CON (272.3 ± 14.3 mL‧min-1, p = 0.003). Mean femoral shear rate was higher in HOT (455.8 ± 25.1 s-1) than CON (405.2 ± 15.8 s-1, p = 0.019). However, there were no differences in any of the investigated genes related to mitochondrial biogenesis (PGC-1α, NRF1, GAPBA, ERRα, TFAM, VEGF) or mitophagy (PINK-1, PARK-2, BNIP-3, BNIP-3L) in response to heat (p > 0.05). These data indicate that heat application alone does not impact the transcriptional response related to mitochondrial homeostasis, suggesting that other factors, in combination with skeletal muscle temperature, are involved with previous observations of altered exercise induced gene expression with heat.
Collapse
Affiliation(s)
- Monica Kwon
- School of Health and Kinesiology, University of Nebraska at Omaha, Omaha, NE 68182, USA
| | - Larry Robins
- School of Health and Kinesiology, University of Nebraska at Omaha, Omaha, NE 68182, USA
| | - Mark L. McGlynn
- School of Health and Kinesiology, University of Nebraska at Omaha, Omaha, NE 68182, USA
| | - Christopher Collins
- School of Health and Kinesiology, University of Nebraska at Omaha, Omaha, NE 68182, USA
| | - Elizabeth J. Pekas
- School of Health and Kinesiology, University of Nebraska at Omaha, Omaha, NE 68182, USA
| | - Song-Young Park
- School of Health and Kinesiology, University of Nebraska at Omaha, Omaha, NE 68182, USA
| | - Dustin Slivka
- School of Health and Kinesiology, University of Nebraska at Omaha, Omaha, NE 68182, USA
- School of Integrative Physiology and Athletic Training, University of Montana, Missoula, MT 59812, USA
| |
Collapse
|
26
|
Wang KC, Yang LY, Lee JE, Wu V, Chen TF, Hsieh ST, Kuo MF. Combination of indirect revascularization and endothelial progenitor cell transplantation improved cerebral perfusion and ameliorated tauopathy in a rat model of bilateral ICA ligation. Stem Cell Res Ther 2022; 13:516. [PMID: 36371197 PMCID: PMC9652785 DOI: 10.1186/s13287-022-03196-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 10/23/2022] [Indexed: 11/15/2022] Open
Abstract
Abstract
Objective
Endothelial progenitor cells (EPCs) contribute to the recovery of neurological function after ischemic stroke. Indirect revascularization has exhibited promising effects in the treatment of cerebral ischemia related to moyamoya disease and intracranial atherosclerotic disease. The role of EPCs in augmenting the revascularization effect is not clear. In this study, we investigated the therapeutic effects of indirect revascularization combined with EPC transplantation in rats with chronic cerebral ischemia.
Methods
Chronic cerebral ischemia was induced by bilateral internal carotid artery ligation (BICAL) in rats, and indirect revascularization by encephalo-myo-synangiosis (EMS) was performed 1 week later. During the EMS procedure, intramuscular injection of EPCs and the addition of stromal cell-derived factor 1 (SDF-1), and AMD3100, an SDF-1 inhibitor, were undertaken, respectively, to investigate their effects on indirect revascularization. Two weeks later, the cortical microcirculation, neuronal damage, and functional outcome were evaluated according to the microvasculature density and partial pressure of brain tissue oxygen (PbtO2), regional blood flow, expression of phosphorylated Tau (pTau), TUNEL staining and the rotarod performance test, respectively.
Results
The cortical microcirculation, according to PbtO2 and regional blood flow, was impaired 3 weeks after BICAL. These impairments were improved by the EMS procedure. The regional blood flow was further increased by the addition of SDF-1 and decreased by the addition of AMD3100. Intramuscular injection of EPCs further increased the regional blood flow as compared with the EMS group. The rotarod test results showed that the functional outcome was best in the EMS combined with EPC injection group. Western blot analysis showed that the EMS combined with EPC treatment group had significantly decreased expressions of phosphorylated Tau and phosphorylated glycogen synthase kinase 3 beta (Y216 of GSK-3β). pTau and TUNEL-positive cells were markedly increased at 3 weeks after BICAL induction. Furthermore, the groups treated with EMS combined with SDF-1 or EPCs exhibited marked decreases in the pTau expression and TUNEL-positive cells, whereas AMD3100 treatment increased TUNEL-positive cells.
Conclusion
The results of this study suggested that indirect revascularization ameliorated the cerebral ischemic changes. EPCs played a key role in augmenting the effect of indirect revascularization in the treatment of chronic cerebral ischemia.
Collapse
|
27
|
Zhang Z, Shen MM, Fu Y. Combination of AIBP, apoA-I, and Aflibercept Overcomes Anti-VEGF Resistance in Neovascular AMD by Inhibiting Arteriolar Choroidal Neovascularization. Invest Ophthalmol Vis Sci 2022; 63:2. [PMID: 36318195 PMCID: PMC9639697 DOI: 10.1167/iovs.63.12.2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Purpose Anti-VEGF resistance represents a major unmet clinical need in the management of choroidal neovascularization (CNV). We have previously reported that a combination of AIBP, apoA-I, and an anti-VEGF antibody overcomes anti-VEGF resistance in laser-induced CNV in old mice in prevention experiments. The purpose of this work is to conduct a more clinically relevant study to assess the efficacy of the combination of AIBP, apoA-I, and aflibercept in the treatment of anti-VEGF resistance of experimental CNV at different time points after laser photocoagulation. Methods To understand the pathobiology of anti-VEGF resistance, we performed comprehensive examinations of the vascular morphology of laser-induced CNV in young mice that are highly responsive to anti-VEGF treatment, and in old mice that are resistant to anti-VEGF therapy by indocyanine green angiography (ICGA), fluorescein angiography (FA), optical coherence tomography (OCT), and Alexa 568 isolectin labeled choroid flatmounts. We examined the efficacy of the combination therapy of AIBP, apoA-I, and aflibercept intravitreally delivered at 2, 4, and 7 days after laser photocoagulation in the treatment of CNV in old mice. Results Laser-induced CNV in young and old mice exhibited cardinal features of capillary and arteriolar CNV, respectively. The combination therapy and the aflibercept monotherapy were equally effective in treating capillary CNV in young mice. In old mice, the combination therapy was effective in treating anti-VEGF resistance by potently inhibiting arteriolar CNV, whereas aflibercept monotherapy was ineffective. Conclusions Combination therapy of AIBP, apoA-I, and aflibercept overcomes anti-VEGF resistance in experimental CNV in old mice by inhibiting arteriolar CNV.
Collapse
Affiliation(s)
- Zhao Zhang
- Cullen Eye Institute, Baylor College of Medicine, Houston, Texas, United States
| | - Megan M. Shen
- Cullen Eye Institute, Baylor College of Medicine, Houston, Texas, United States
| | - Yingbin Fu
- Cullen Eye Institute, Baylor College of Medicine, Houston, Texas, United States
| |
Collapse
|
28
|
McCrary MR, Jiang MQ, Jesson K, Gu X, Logun MT, Wu A, Gonsalves N, Karumbaiah L, Yu SP, Wei L. Glycosaminoglycan scaffolding and neural progenitor cell transplantation promotes regenerative immunomodulation in the mouse ischemic brain. Exp Neurol 2022; 357:114177. [PMID: 35868359 PMCID: PMC10066865 DOI: 10.1016/j.expneurol.2022.114177] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 07/04/2022] [Accepted: 07/16/2022] [Indexed: 02/08/2023]
Abstract
Ischemic stroke is a leading cause of morbidity and mortality, with limited treatments that can facilitate brain regeneration. Neural progenitor cells (NPCs) hold promise for replacing tissue lost to stroke, and biomaterial approaches may improve their efficacy to overcome hurdles in clinical translation. The immune response and its role in stroke pathogenesis and regeneration may interplay with critical mechanisms of stem cell and biomaterial therapies. Cellular therapy can modulate the immune response to reduce toxic neuroinflammation early after ischemia. However, few studies have attempted to harness the regenerative effects of neuroinflammation to augment recovery. Our previous studies demonstrated that intracerebrally transplanted NPCs encapsulated in a chondroitin sulfate-A hydrogel (CS-A + NPCs) can improve vascular regeneration after stroke. In this paper, we found that CS-A + NPCs affect the microglia/macrophage response to promote a regenerative phenotype following stroke in mice. Following transplantation, PPARγ-expressing microglia/macrophages, and MCP-1 and IL-10 protein levels are enhanced. Secreted immunomodulatory factor expression of other factors was altered compared to NPC transplantation alone. Post-stroke depression-like behavior was reduced following cellular and material transplantation. Furthermore, we showed in cultures that microglia/macrophages encapsulated in CS-A had increased expression of angiogenic and arteriogenic mediators. Neutralization with anti-IL-10 antibody negated these effects in vitro. Cumulatively, this work provides a framework for understanding the mechanisms by which immunomodulatory biomaterials can enhance the regenerative effects of cellular therapy for ischemic stroke and other brain injuries.
Collapse
Affiliation(s)
- Myles R McCrary
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Michael Q Jiang
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA; Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, GA, USA
| | - Kaleena Jesson
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Xiaohuan Gu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA; Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, GA, USA
| | - Meghan T Logun
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA; Division of Neuroscience, Biomedical and Translational Sciences Institute, University of Georgia, Athens, GA, USA
| | - Anika Wu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Nathan Gonsalves
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA; Division of Neuroscience, Biomedical and Translational Sciences Institute, University of Georgia, Athens, GA, USA
| | - Lohitash Karumbaiah
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA; Division of Neuroscience, Biomedical and Translational Sciences Institute, University of Georgia, Athens, GA, USA; Department of Animal and Dairy Science, College of Agriculture and Environmental Sciences, University of Georgia, Athens, GA, USA
| | - Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA; Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, GA, USA
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
29
|
Shin HS, Thakore A, Tada Y, Pedroza AJ, Ikeda G, Chen IY, Chan D, Jaatinen KJ, Yajima S, Pfrender EM, Kawamura M, Yang PC, Wu JC, Appel EA, Fischbein MP, Woo YJ, Shudo Y. Angiogenic stem cell delivery platform to augment post-infarction neovasculature and reverse ventricular remodeling. Sci Rep 2022; 12:17605. [PMID: 36266453 PMCID: PMC9584918 DOI: 10.1038/s41598-022-21510-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 09/28/2022] [Indexed: 01/13/2023] Open
Abstract
Many cell-based therapies are challenged by the poor localization of introduced cells and the use of biomaterial scaffolds with questionable biocompatibility or bio-functionality. Endothelial progenitor cells (EPCs), a popular cell type used in cell-based therapies due to their robust angiogenic potential, are limited in their therapeutic capacity to develop into mature vasculature. Here, we demonstrate a joint delivery of human-derived endothelial progenitor cells (EPC) and smooth muscle cells (SMC) as a scaffold-free, bi-level cell sheet platform to improve ventricular remodeling and function in an athymic rat model of myocardial infarction. The transplanted bi-level cell sheet on the ischemic heart provides a biomimetic microenvironment and improved cell-cell communication, enhancing cell engraftment and angiogenesis, thereby improving ventricular remodeling. Notably, the increased density of vessel-like structures and upregulation of biological adhesion and vasculature developmental genes, such as Cxcl12 and Notch3, particularly in the ischemic border zone myocardium, were observed following cell sheet transplantation. We provide compelling evidence that this SMC-EPC bi-level cell sheet construct can be a promising therapy to repair ischemic cardiomyopathy.
Collapse
Affiliation(s)
- Hye Sook Shin
- Department of Cardiothoracic Surgery, Falk Cardiovascular Research Center, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, USA
| | - Akshara Thakore
- Department of Cardiothoracic Surgery, Falk Cardiovascular Research Center, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, USA
| | - Yuko Tada
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, USA
| | - Albert J Pedroza
- Department of Cardiothoracic Surgery, Falk Cardiovascular Research Center, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, USA
| | - Gentaro Ikeda
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, USA
| | - Ian Y Chen
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, USA
| | - Doreen Chan
- Department of Chemistry, Department of Materials Science & Engineering, Stanford University, Stanford University, Stanford, USA
| | - Kevin J Jaatinen
- Department of Cardiothoracic Surgery, Falk Cardiovascular Research Center, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Shin Yajima
- Department of Cardiothoracic Surgery, Falk Cardiovascular Research Center, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, USA
| | - Eric M Pfrender
- Department of Cardiothoracic Surgery, Falk Cardiovascular Research Center, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, USA
| | - Masashi Kawamura
- Department of Cardiothoracic Surgery, Falk Cardiovascular Research Center, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Phillip C Yang
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, USA
| | - Joseph C Wu
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, USA
| | - Eric A Appel
- Department of Materials Science & Engineering, Department of Bioengineering, Department of Pediatric (Endocrinology), Stanford University, Stanford, USA
| | - Michael P Fischbein
- Department of Cardiothoracic Surgery, Falk Cardiovascular Research Center, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, USA
| | - YJoseph Woo
- Department of Cardiothoracic Surgery, Falk Cardiovascular Research Center, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, USA
| | - Yasuhiro Shudo
- Department of Cardiothoracic Surgery, Falk Cardiovascular Research Center, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, USA.
| |
Collapse
|
30
|
He L, Lu H, Chu J, Qin X, Gao J, Chen M, Weinstein LS, Yang J, Zhang Q, Zhang C, Zhang W. Endothelial G protein stimulatory α-subunit is a critical regulator of post-ischemic angiogenesis. Front Cardiovasc Med 2022; 9:941946. [PMID: 35958407 PMCID: PMC9358140 DOI: 10.3389/fcvm.2022.941946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/05/2022] [Indexed: 11/13/2022] Open
Abstract
Post-ischemic angiogenesis is a vital pathophysiological process in diseases such as peripheral arterial disease (PAD), heart ischemia, and diabetic retinopathy. The molecular mechanisms of post-ischemic angiogenesis are complicated and not fully elucidated. The G protein stimulatory alpha subunit (Gsα) is essential for hormone-stimulated cyclic adenosine monophosphate (cAMP) production and is an important regulator for many physiological processes. In the present study, we investigated the role of endothelial Gsα in post-ischemic angiogenesis by generating adult mice with endothelial-specific Gsα deficiency (GsαECKO). GsαECKO mice had impaired blood flow recovery after hind limb ischemic injury, and reduced neovascularization in allograft transplanted tumors. Mechanically, Gsα could regulate the expression of angiogenic factor with G patch and FHA domains 1 (AGGF1) through cAMP/CREB pathway. AGGF1 plays a key role in angiogenesis and regulates endothelial cell proliferation as well as migration. Knockdown of CREB or mutation of the CRE site on the AGGF1 promoter led to reduced AGGF1 promoter activity. In addition, knockdown of AGGF1 reduced the proangiogenic effect of Gsα in endothelial cells, and overexpression of AGGF1 reversed the impaired angiogenesis in GsαECKO mice in vivo. The finding may prove useful in designing new therapeutic targets for treatments of post-ischemic angiogenesis-related diseases.
Collapse
Affiliation(s)
- Lifan He
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hanlin Lu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Cardiovascular Disease Research Center of Shandong First Medical University, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jianying Chu
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, China
| | - Xiaoteng Qin
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jiangang Gao
- School of Life Sciences and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, China
| | - Min Chen
- Metabolic Diseases Branch, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Lee S. Weinstein
- Metabolic Diseases Branch, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Jianmin Yang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qunye Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Cheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wencheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Cardiovascular Disease Research Center of Shandong First Medical University, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
- *Correspondence: Wencheng Zhang,
| |
Collapse
|
31
|
Li J, Li R, Wu X, Zheng C, Shiu PHT, Rangsinth P, Lee SMY, Leung GPH. An Update on the Potential Application of Herbal Medicine in Promoting Angiogenesis. Front Pharmacol 2022; 13:928817. [PMID: 35928282 PMCID: PMC9345329 DOI: 10.3389/fphar.2022.928817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Angiogenesis, the formation of new capillaries from pre-existing vascular networks, plays an important role in many physiological and pathological processes. The use of pro-angiogenic agents has been proposed as an attractive approach for promoting wound healing and treating vascular insufficiency-related problems, such as ischemic heart disease and stroke, which are the leading causes of death worldwide. Traditional herbal medicine has a long history; however, there is still a need for more in-depth studies and evidence-based confirmation from controlled and validated trials. Many in vitro and in vivo studies have reported that herbal medicines and their bioactive ingredients exert pro-angiogenic activity. The most frequently studied pro-angiogenic phytochemicals include ginsenosides from Panax notoginseng, astragalosides and calycosin from Radix Astragali, salvianolic acid B from Salvia miltiorrhiza, paeoniflorin from Radix Paeoniae, ilexsaponin A1 from Ilex pubescens, ferulic acid from Angelica sinensis, and puerarin from Radix puerariae. This review summarizes the progress in research on these phytochemicals, particularly those related to pro-angiogenic mechanisms and applications in ischemic diseases, tissue repair, and wound healing. In addition, an outline of their limitations and challenges during drug development is presented.
Collapse
Affiliation(s)
- Jingjing Li
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Renkai Li
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Xiaoping Wu
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Chengwen Zheng
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Polly Ho-Ting Shiu
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Panthakarn Rangsinth
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Simon Ming-Yuen Lee
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Taipa Macao SAR, China
| | - George Pak-Heng Leung
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- *Correspondence: George Pak-Heng Leung,
| |
Collapse
|
32
|
The Interplay of Endothelial P2Y Receptors in Cardiovascular Health: From Vascular Physiology to Pathology. Int J Mol Sci 2022; 23:ijms23115883. [PMID: 35682562 PMCID: PMC9180512 DOI: 10.3390/ijms23115883] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/19/2022] [Accepted: 05/23/2022] [Indexed: 12/19/2022] Open
Abstract
The endothelium plays a key role in blood vessel health. At the interface of the blood, it releases several mediators that regulate local processes that protect against the development of cardiovascular disease. In this interplay, there is increasing evidence for a role of extracellular nucleotides and endothelial purinergic P2Y receptors (P2Y-R) in vascular protection. Recent advances have revealed that endothelial P2Y1-R and P2Y2-R mediate nitric oxide-dependent vasorelaxation as well as endothelial cell proliferation and migration, which are processes involved in the regeneration of damaged endothelium. However, endothelial P2Y2-R, and possibly P2Y1-R, have also been reported to promote vascular inflammation and atheroma development in mouse models, with endothelial P2Y2-R also being described as promoting vascular remodeling and neointimal hyperplasia. Interestingly, at the interface with lipid metabolism, P2Y12-R has been found to trigger HDL transcytosis through endothelial cells, a process known to be protective against lipid deposition in the vascular wall. Better characterization of the role of purinergic P2Y-R and downstream signaling pathways in determination of the endothelial cell phenotype in healthy and pathological environments has clinical potential for the prevention and treatment of cardiovascular diseases.
Collapse
|
33
|
Predicting lesion shrinkage in eyes with myopic choroidal neovascularization from features on optical coherence tomography angiography. Retina 2022; 42:1665-1672. [PMID: 35594547 DOI: 10.1097/iae.0000000000003526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE To identify baseline morphological predictors of lesion shrinkage in eyes with myopic choroidal neovascularization (mCNV) treated with anti-vascular endothelial growth factor (VEGF). METHODS This retrospective study included 46 eyes (41 consecutive patients) with active mCNV receiving anti-VEGF treatment. Optical coherence tomography angiography (OCTA) was performed at baseline and 1 year after treatment. Quantitative features were obtained from OCTA images using AngioTool software. Eyes were classified as "high-shrinkage" or "low-shrinkage" according to the median relative change in lesion area. Baseline quantitative morphological features associated with mCNV shrinkage were identified in univariate and multivariate analyses. RESULTS The mCNV area was significantly smaller after 1 year (P=0.013), with a median relative change of -16.5%. The relative change in mCNV area was -48.3% in high-shrinkage eyes (n=23) and -5.2% in low-shrinkage eyes (n=23). High-shrinkage eyes had a smaller mCNV area (P=0.013), shorter total vessel length (P=0.023), and higher endpoint density (P<0.001). Multivariate analysis showed significant associations of high shrinkage with endpoint density (β=-0.037, P=0.043) and previous anti-VEGF treatment (β=0.216, P=0.029). CONCLUSION Morphological features of neovascularization detected by OCTA can predict lesion shrinkage in eyes with mCNV receiving anti-VEGF therapy. Higher endpoint density contributed to shrinkage, particularly of treatment-naive lesions.
Collapse
|
34
|
O'Hare M, Arboleda-Velasquez JF. Notch Signaling in Vascular Endothelial and Mural Cell Communications. Cold Spring Harb Perspect Med 2022; 12:a041159. [PMID: 35534207 PMCID: PMC9435572 DOI: 10.1101/cshperspect.a041159] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The Notch signaling pathway is a highly versatile and evolutionarily conserved mechanism with an important role in cell fate determination. Notch signaling plays a vital role in vascular development, regulating several fundamental processes such as angiogenesis, arterial/venous differentiation, and mural cell investment. Aberrant Notch signaling can result in severe vascular phenotypes as observed in cerebral autosomal-dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) and Alagille syndrome. It is known that vascular endothelial cells and mural cells interact to regulate vessel formation, cell maturation, and stability of the vascular network. Defective endothelial-mural cell interactions are a common phenotype in diseases characterized by impaired vascular integrity. Further refinement of the role of Notch signaling in the vascular junctions will be critical to attempts to modulate Notch in the context of human vascular disease. In this review, we aim to consolidate and summarize our current understanding of Notch signaling in the vascular endothelial and mural cells during development and in the adult vasculature.
Collapse
Affiliation(s)
- Michael O'Hare
- Department of Ophthalmology at Harvard Medical School, Schepens Eye Research Institute of Mass Eye and Ear, Boston, Massachusetts 02114, USA
| | - Joseph F Arboleda-Velasquez
- Department of Ophthalmology at Harvard Medical School, Schepens Eye Research Institute of Mass Eye and Ear, Boston, Massachusetts 02114, USA
| |
Collapse
|
35
|
Xu Y, Ward AD, Goldman D, Yin H, Arpino JM, Nong Z, Lee JJ, O'Neil C, Pickering JG. Arteriolar dysgenesis in ischemic, regenerating skeletal muscle revealed by automated micro-morphometry, computational modeling, and perfusion analysis. Am J Physiol Heart Circ Physiol 2022; 323:H38-H48. [PMID: 35522554 DOI: 10.1152/ajpheart.00010.2022] [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: 11/22/2022]
Abstract
Rebuilding the local vasculature is central to restoring the health of muscles subjected to ischemic injury. Arteriogenesis yields remodeled collateral arteries that circumvent the obstruction, and angiogenesis produces capillaries to perfuse the regenerating myofibers. However, the vital intervening network of arterioles that feed the regenerated capillaries is poorly understood and an investigative challenge. We used machine learning and automated micro-morphometry to quantify the arteriolar landscape in distal hindlimb muscles in mice that have regenerated after femoral artery excision. Assessment of 1546 arteriolar sections revealed a striking (> 2-fold) increase in arteriolar density in regenerated muscle 14 and 28 days after ischemic injury. Lumen caliber was initially similar to that of control arterioles but after 4 weeks lumen area was reduced by 46%. In addition, the critical smooth muscle layer was attenuated throughout the arteriolar network, across a 150 to 5 µm diameter range. To understand the consequences of the reshaped distal hindlimb arterioles, we undertook computational flow modeling which revealed blunted flow augmentation. Moreover, impaired flow reserve was confirmed in vivo by laser Doppler analyses of flow in response to directly applied sodium nitroprusside. Thus, in hindlimb muscles regenerating after ischemic injury, the arteriolar network is amplified, inwardly remodels, and is diffusely under-muscularized. These defects and the associated flow restraints could contribute to the deleterious course of peripheral artery disease and merit attention when considering therapeutic innovations.
Collapse
Affiliation(s)
- Yiwen Xu
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada.,Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada
| | - Aaron D Ward
- Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada
| | - Daniel Goldman
- Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada
| | - Hao Yin
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - John-Michael Arpino
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada.,Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada
| | - Zengxuan Nong
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Jason J Lee
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada.,Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada.,Department of Medicine, University of Western Ontario, London, Ontario, Canada
| | - Caroline O'Neil
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - J Geoffrey Pickering
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada.,Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada.,Department of Biochemistry, University of Western Ontario, London, Ontario, Canada.,Department of Medicine, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
36
|
Bao XY, Fan YN, Wang QN, Wang XP, Yang RM, Zou ZX, Zhang Q, Li DS, Duan L, Yu XG. The Potential Mechanism Behind Native and Therapeutic Collaterals in Moyamoya. Front Neurol 2022; 13:861184. [PMID: 35557620 PMCID: PMC9086844 DOI: 10.3389/fneur.2022.861184] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/23/2022] [Indexed: 11/13/2022] Open
Abstract
Background and Purpose To explore the genetic basis and molecular mechanism of native arteriogenesis and therapeutic synangiosis in moyamoya disease (MMD). Methods An angiography-based study using patients from a prospective trial of encephaloduroarteriosynangiosis (EDAS) surgery was performed. The spontaneous collaterals grades were evaluated according to the system described by a new grading system. Blood samples were collected from all the recruited patients before EDAS and during the second hospitalization 3 months post-EDAS. We performed Boolean analysis using a combination of specific cell surface markers of CD34briCD133+CD45dimKDR+. Genotyping of p.R4810K was also performed. The correlation of age, sex, initial symptoms at diagnosis, collateral grade, Suzuki stages, the RNF213 genotype, time to peak (TTP), and endothelial progenitor cell (EPC) count with good collateral circulation was evaluated. Results Eighty-five patients with MMD were included in this study. The mutation rate of RNF213 p.R4810K in our study was 25.9% (22/85). The heterozygous mutations were occurred significantly more frequently in the cases that were presented with infarction, worse neurological status, severe posterior cerebral artery (PCA) stenosis, and longer TTP delay. Further, the heterozygous mutations occurred significantly more frequently in the poor collateral stage group. Lower grades were significantly correlated with severe ischemia symptoms, worse neurological status, and a longer TTP delay. The post-operative angiographic findings showed that a good Matsushima grade was correlated with heterozygous mutations, a lower collateral stage, and a longer TTP delay. The CD34briCD133+CD45dimKDR+ cell count in patients 3 months post-EDAS was significantly higher as compared to the count before EDAS in the good Matsushima grade group. However, this change was not observed in the poor Matsushima grade group. Conclusions These data imply that mutations of RNF213 p.R4810K affect the establishment of spontaneous collateral circulation, and EPCs are involved in the process of formation of new EDAS collaterals.
Collapse
Affiliation(s)
- Xiang-Yang Bao
- Department of Neurosurgery, Chinese People's Liberation Army of China (PLA) General Hospital, Beijing, China
- Chinese PLA Medical School, Beijing, China
| | - Yan-Na Fan
- Department of Radiation Oncology, The Fifth Medical Center of Chinese PLA General Hospital (Former 307th Hospital of the PLA), Beijing, China
| | - Qian-Nan Wang
- Department of Neurosurgery, Chinese People's Liberation Army of China (PLA) General Hospital, Beijing, China
| | - Xiao-Peng Wang
- Department of Neurosurgery, Chinese People's Liberation Army of China (PLA) General Hospital, Beijing, China
| | - Ri-Miao Yang
- Department of Neurosurgery, Chinese People's Liberation Army of China (PLA) General Hospital, Beijing, China
| | - Zheng-Xing Zou
- Department of Neurosurgery, Chinese People's Liberation Army of China (PLA) General Hospital, Beijing, China
| | - Qian Zhang
- Department of Neurosurgery, Chinese People's Liberation Army of China (PLA) General Hospital, Beijing, China
| | - De-Sheng Li
- Department of Neurosurgery, Chinese People's Liberation Army of China (PLA) General Hospital, Beijing, China
| | - Lian Duan
- Department of Neurosurgery, Chinese People's Liberation Army of China (PLA) General Hospital, Beijing, China
- *Correspondence: Lian Duan
| | - Xin-Guang Yu
- Department of Neurosurgery, Chinese People's Liberation Army of China (PLA) General Hospital, Beijing, China
- Chinese PLA Medical School, Beijing, China
- Xin-Guang Yu
| |
Collapse
|
37
|
Schawe L, Raude B, Carstens JC, Hinterseher I, Hein RD, Omran S, Berger G, Hering NA, Buerger M, Greiner A, Frese JP. Effect of Revascularization on Intramuscular Vascular Endothelial Growth Factor Levels in Peripheral Arterial Disease. Biomedicines 2022; 10:471. [PMID: 35203679 PMCID: PMC8962418 DOI: 10.3390/biomedicines10020471] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/05/2022] [Accepted: 02/10/2022] [Indexed: 02/01/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) is a potent driver of angiogenesis, which may help to relieve ischemia in peripheral arterial disease (PAD). We aimed to investigate the role of intramuscular VEGF in ischemic and non-ischemic skeletal muscle in PAD patients before and after surgical or endovascular revascularization and different stages of PAD. Biopsies of the gastrocnemius and vastus muscles from twenty PAD patients with stenosis or occlusion of the superficial femoral artery were obtained both during revascularization and 8 weeks postoperatively. The gastrocnemius muscle was considered ischemic, while vastus muscle biopsies served as intraindividual controls. The levels of vascular endothelial growth factor in muscle lysates were then determined by ELISA. Preoperative VEGF levels were significantly higher in ischemic muscles compared to the controls (98.07 ± 61.96 pg/mL vs. 55.50 ± 27.33 pg/mL, p = 0.004). Postoperative values decreased significantly (p = 0.010) to 54.83 ± 49.60 pg/mL in gastrocnemius biopsies. No significant change was observed in vastus muscle biopsies, with mean postoperative VEGF values found at 54.16 ± 40.66 pg/mL. Since all patients still had indications for revascularization, impairment of angiogenesis mechanisms can be assumed. More research about angiogenesis in PAD is needed with the ultimate goal to improve conservative treatment.
Collapse
Affiliation(s)
- Larissa Schawe
- Department of Vascular Surgery, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (B.R.); (J.C.C.); (I.H.); (S.O.); (M.B.); (A.G.); (J.P.F.)
| | - Ben Raude
- Department of Vascular Surgery, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (B.R.); (J.C.C.); (I.H.); (S.O.); (M.B.); (A.G.); (J.P.F.)
| | - Jan Christoph Carstens
- Department of Vascular Surgery, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (B.R.); (J.C.C.); (I.H.); (S.O.); (M.B.); (A.G.); (J.P.F.)
| | - Irene Hinterseher
- Department of Vascular Surgery, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (B.R.); (J.C.C.); (I.H.); (S.O.); (M.B.); (A.G.); (J.P.F.)
- Department of Vascular Surgery, Medizinische Hochschule Brandenburg Theodor Fontane, Ruppiner Kliniken—University Hospital, 16816 Neuruppin, Germany
| | - Raphael Donatus Hein
- Department of Anaesthesiology and Intensive Care Medicine, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany;
| | - Safwan Omran
- Department of Vascular Surgery, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (B.R.); (J.C.C.); (I.H.); (S.O.); (M.B.); (A.G.); (J.P.F.)
| | - Gilles Berger
- Microbiology, Bioorganic & Macromolecular Chemistry, Faculté de Pharmacie, Université Libre de Bruxelles (ULB), Bd du Triomphe, 1050 Brussels, Belgium;
| | - Nina A. Hering
- Department of General and Visceral Surgery, Campus Benjamin Franklin, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany;
| | - Matthias Buerger
- Department of Vascular Surgery, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (B.R.); (J.C.C.); (I.H.); (S.O.); (M.B.); (A.G.); (J.P.F.)
| | - Andreas Greiner
- Department of Vascular Surgery, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (B.R.); (J.C.C.); (I.H.); (S.O.); (M.B.); (A.G.); (J.P.F.)
| | - Jan Paul Frese
- Department of Vascular Surgery, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (B.R.); (J.C.C.); (I.H.); (S.O.); (M.B.); (A.G.); (J.P.F.)
| |
Collapse
|
38
|
Eton D, Zhou G, He TC, Bartholomew A, Patil R. Filgrastim, fibrinolysis, and neovascularization. J Tissue Eng Regen Med 2022; 16:496-510. [PMID: 35175691 PMCID: PMC9302657 DOI: 10.1002/term.3284] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 12/17/2021] [Accepted: 01/11/2022] [Indexed: 12/11/2022]
Abstract
Segmental recanalization of chronically occluded arteries was observed in patients with chronic limb-threatening ischemia (CLTI) treated with Filgrastim, a granulocyte colony stimulating factor, every 72 h for up to a month, and an infra-geniculate programmed compression pump (PCP) for 3 h daily. Molecular evidence for fibrinolysis and neovascularization was sought. CLTI patients were treated with PCP alone (N = 19), or with Filgrastim and PCP (N = 8 and N = 6, at two institutions). Enzyme-Linked Immunosorbent Assay was used to measure the plasma concentration of plasmin and of fibrin degradation products (FDP), and the serum concentration of proteins associated with neovascularization. In the PCP-alone group, blood was sampled on Day 1 (baseline) and after 30 days of daily PCP. In the Filgrastim and PCP group, blood was drawn on Day 1, and 1 day after the 5th and the 10th Filgrastim doses. Each blood draw occurred before and after 2 h of supervised PCP. Significant (p < 0.01) PCP independent increases in the plasma concentration of plasmin (>10-fold) and FDP (>5-fold) were observed 1 day after both the 5th and the 10th Filgrastim doses, compared to Day 1. Significant (p < 0.05) increases in the concentration of pro-angiogenic proteins (e.g., HGF, MMP-9, VEGF A) were also observed. Filgrastim at this novel dosimetry induced fibrinolysis without causing acute hemorrhage, in addition to inducing a pro-angiogenic milieu conducive to NV. Further clinical testing is warranted at this novel dosimetry in CLTI, as well as in other chronically ischemic tissue beds. Trial registration. https://clinicaltrials.gov/ct2/show/NCT02802852.
Collapse
Affiliation(s)
- Darwin Eton
- Department of Surgery, University of Illinois Chicago, Chicago, Illinois, USA
| | - Guolin Zhou
- GCIS, University of Chicago, Chicago, Illinois, USA
| | - Tong-Chuan He
- Department of Orthopedic Surgery and Rehabilitation Medicine, University of Chicago, Chicago, Illinois, USA
| | - Amelia Bartholomew
- Department of Surgery, College of Medicine Research, University of Illinois Chicago, Chicago, Illinois, USA
| | - Rachana Patil
- Department of Pediatrics, Division of Stem Cell Transplant and Regenerative Medicine, Stanford University, Palo Alto, California, USA
| |
Collapse
|
39
|
Goonoo N. Tunable Biomaterials for Myocardial Tissue Regeneration: Promising New Strategies for Advanced Biointerface Control and Improved Therapeutic Outcomes. Biomater Sci 2022; 10:1626-1646. [DOI: 10.1039/d1bm01641e] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Following myocardial infarction (MI) and the natural healing process, the cardiac mechanostructure changes significantly leading to reduced contractile ability and putting additional pressure on the heart muscle thereby increasing the...
Collapse
|
40
|
Bolandi SM, Abdolmaleki Z, Assarehzadegan MA. Anti-angiogenic Properties of Bevacizumab Improve Respiratory System Inflammation in Ovalbumin-Induced Rat Model of Asthma. Inflammation 2021; 44:2463-2475. [PMID: 34420156 PMCID: PMC8380193 DOI: 10.1007/s10753-021-01516-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 06/16/2021] [Accepted: 07/07/2021] [Indexed: 02/07/2023]
Abstract
Studies on the bronchial vascular bed have revealed that the number of blood vessels in the lamina propria and under the mucosa of the lung tissue increases in patients suffering from mild to severe asthma. Thus, in this study, a new strategy was employed in respiratory system disorders by angiogenesis inhibition in an ovalbumin (OVA)-induced rat model of asthma. Twenty-one male Wistar albino rats, 8 weeks old, were randomly divided into three groups (n = 7 in each group), including (1) control group, (2) OVA-treated group, and (3) OVA + Bmab (bevacizumab drug). On days 1 and 8, 1 mg of OVA and aluminum hydroxide in sterile phosphate-buffered saline (PBS) were intraperitoneally injected to rats in groups 2 and 3. The control group was only subject to intraperitoneal injection of saline on days 1 and 8. One week after the last injection, the rats (groups 2 and 3) were exposed to OVA inhalation for 30 min at 2-day intervals from days 15 to 25. After sensitization and challenge with OVA, the OVA + Bmab group (group 3) were treated with a 5 mg/kg bevacizumab drug. Genes and protein expression of IL-1β and TNF-α and the expression of vascular endothelial growth factor (VEGF) protein were assessed by real-time PCR and immunohistochemistry respectively, in lung tissue. OVA exposure increased mucosal secretion and inflammatory cell populations in lung tissue and OVA-specific IgE level in serum. Also, VEGF and cytokine factor expression were significantly elevated in the OVA-induced asthma model (p ≤ 0.05). However, rats in OVA + Bmab group showed significantly a decrease in VEGF and IL-1β and TNF-α genes as well as proteins (p ≤ 0.05). The results showed that bevacizumab efficiently diminished bronchial inflammation via downregulation of VEGF expression, followed by inflammatory cells population and cytokines reduction. Angiogenesis inhibition in rats with induced asthma not only suppresses the inflammatory process through blocking VEGF expression but also inhibits the development of new blood vessels and progressing asthmatic attacks.
Collapse
Affiliation(s)
| | - Zohreh Abdolmaleki
- Department of Pharmacology, Karaj Branch, Islamic Azad University, Karaj, Iran.
- Department of Pharmacology, Karaj Branch, Islamic Azad University, Karaj, Iran.
| | - Mohammad-Ali Assarehzadegan
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
41
|
Kübler M, Götz P, Braumandl A, Beck S, Ishikawa-Ankerhold H, Deindl E. Impact of C57BL/6J and SV-129 Mouse Strain Differences on Ischemia-Induced Postnatal Angiogenesis and the Associated Leukocyte Infiltration in a Murine Hindlimb Model of Ischemia. Int J Mol Sci 2021; 22:ijms222111795. [PMID: 34769229 PMCID: PMC8584150 DOI: 10.3390/ijms222111795] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/23/2021] [Accepted: 10/27/2021] [Indexed: 12/11/2022] Open
Abstract
Strain-related differences in arteriogenesis in inbred mouse strains have already been studied excessively. However, these analyses missed evaluating the mouse strain-related differences in ischemia-induced angiogenic capacities. With the present study, we wanted to shed light on the different angiogenic potentials and the associated leukocyte infiltration of C57BL/6J and SV-129 mice to facilitate the comparison of angiogenesis-related analyses between these strains. For the induction of angiogenesis, we ligated the femoral artery in 8-12-week-old male C57BL/6J and SV-129 mice and performed (immuno-) histological analyses on the ischemic gastrocnemius muscles collected 24 h or 7 days after ligation. As evidenced by hematoxylin and eosin staining, C57BL/6J mice showed reduced tissue damage but displayed an increased capillary-to-muscle fiber ratio and an elevated number of proliferating capillaries (CD31+/BrdU+ cells) compared to SV-129 mice, thus showing improved angiogenesis. Regarding the associated leukocyte infiltration, we found increased numbers of neutrophils (MPO+ cells), NETs (MPO+/CitH3+/DAPI+), and macrophages (CD68+ cells) in SV-129 mice, whereas macrophage polarization (MRC1- vs. MRC1+) and total leukocyte infiltration (CD45+ cells) did not differ between the mouse strains. In summary, we show increased ischemia-induced angiogenic capacities in C57BL/6J mice compared to SV-129 mice, with the latter showing aggravated tissue damage, inflammation, and impaired angiogenesis.
Collapse
Affiliation(s)
- Matthias Kübler
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; (M.K.); (P.G.); (A.B.); (S.B.); (H.I.-A.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Philipp Götz
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; (M.K.); (P.G.); (A.B.); (S.B.); (H.I.-A.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Anna Braumandl
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; (M.K.); (P.G.); (A.B.); (S.B.); (H.I.-A.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Sebastian Beck
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; (M.K.); (P.G.); (A.B.); (S.B.); (H.I.-A.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Hellen Ishikawa-Ankerhold
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; (M.K.); (P.G.); (A.B.); (S.B.); (H.I.-A.)
- Department of Internal Medicine I, Faculty of Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Elisabeth Deindl
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; (M.K.); (P.G.); (A.B.); (S.B.); (H.I.-A.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
- Correspondence: ; Tel.: +49-(0)-89-2180-76504
| |
Collapse
|
42
|
Plicosepalus acacia Extract and Its Major Constituents, Methyl Gallate and Quercetin, Potentiate Therapeutic Angiogenesis in Diabetic Hind Limb Ischemia: HPTLC Quantification and LC-MS/MS Metabolic Profiling. Antioxidants (Basel) 2021; 10:antiox10111701. [PMID: 34829572 PMCID: PMC8614836 DOI: 10.3390/antiox10111701] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/21/2021] [Accepted: 10/25/2021] [Indexed: 11/23/2022] Open
Abstract
Plicosepalus acacia (Fam. Loranthaceae) has been reported to possess hypoglycemic, antioxidant, antimicrobial, and anti-inflammatory effects. Liquid chromatography combined with tandem mass spectrometry (LC-MS/MS) analysis revealed the presence of a high content of polyphenolic compounds that are attributed to the therapeutic effects of the crude extract. In addition, methyl gallate and quercetin were detected as major phytomedicinal agents at concentrations of 1.7% and 0.062 g%, respectively, using high-performance thin layer chromatography (HPTLC). The present study investigated the effect of the P. acacia extract and its isolated compounds, methyl gallate and quercetin, on hind limb ischemia induced in type 1 diabetic rats. Histopathological examination revealed that treatment with P. acacia extract, methyl gallate, and quercetin decreased degenerative changes and inflammation in the ischemic muscle. Further biochemical assessment of the hind limb tissue showed decreased oxidative stress, increased levels of nitric oxide and endothelial nitric oxide synthase (eNOS), and enhancement of the levels of heme oxygenase-1 (HO-1) and vascular endothelial growth factor (VEGF) in the groups treated with methyl gallate and quercetin. Expression levels of hypoxia inducible factor-1 alpha (HIF-1α), VEGF, fibroblast growth factor-2 (FGF-2), and miR-146a were upregulated in the muscle tissue of methyl gallate- and quercetin-treated groups along with downregulation of nuclear factor kappa B (NF-κB). In conclusion, P. acacia extract and its isolated compounds, methyl gallate and quercetin, mediated therapeutic angiogenesis in diabetic hind limb ischemia.
Collapse
|
43
|
Kübler M, Beck S, Peffenköver LL, Götz P, Ishikawa-Ankerhold H, Preissner KT, Fischer S, Lasch M, Deindl E. The Absence of Extracellular Cold-Inducible RNA-Binding Protein (eCIRP) Promotes Pro-Angiogenic Microenvironmental Conditions and Angiogenesis in Muscle Tissue Ischemia. Int J Mol Sci 2021; 22:ijms22179484. [PMID: 34502391 PMCID: PMC8431021 DOI: 10.3390/ijms22179484] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/27/2021] [Accepted: 08/30/2021] [Indexed: 12/11/2022] Open
Abstract
Extracellular Cold-inducible RNA-binding protein (eCIRP), a damage-associated molecular pattern, is released from cells upon hypoxia and cold-stress. The overall absence of extra- and intracellular CIRP is associated with increased angiogenesis, most likely induced through influencing leukocyte accumulation. The aim of the present study was to specifically characterize the role of eCIRP in ischemia-induced angiogenesis together with the associated leukocyte recruitment. For analyzing eCIRPs impact, we induced muscle ischemia via femoral artery ligation (FAL) in mice in the presence or absence of an anti-CIRP antibody and isolated the gastrocnemius muscle for immunohistological analyses. Upon eCIRP-depletion, mice showed increased capillary/muscle fiber ratio and numbers of proliferating endothelial cells (CD31+/CD45−/BrdU+). This was accompanied by a reduction of total leukocyte count (CD45+), neutrophils (MPO+), neutrophil extracellular traps (NETs) (MPO+CitH3+), apoptotic area (ascertained via TUNEL assay), and pro-inflammatory M1-like polarized macrophages (CD68+/MRC1−) in ischemic muscle tissue. Conversely, the number of regenerative M2-like polarized macrophages (CD68+/MRC1+) was elevated. Altogether, we observed that eCIRP depletion similarly affected angiogenesis and leukocyte recruitment as described for the overall absence of CIRP. Thus, we propose that eCIRP is mainly responsible for modulating angiogenesis via promoting pro-angiogenic microenvironmental conditions in muscle ischemia.
Collapse
Affiliation(s)
- Matthias Kübler
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; (M.K.); (S.B.); (P.G.); (H.I.-A.); (M.L.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig- Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Sebastian Beck
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; (M.K.); (S.B.); (P.G.); (H.I.-A.); (M.L.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig- Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Lisa Lilian Peffenköver
- Department of Biochemistry, Faculty of Medicine, Justus Liebig University, 35392 Giessen, Germany; (L.L.P.); (K.T.P.); (S.F.)
| | - Philipp Götz
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; (M.K.); (S.B.); (P.G.); (H.I.-A.); (M.L.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig- Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Hellen Ishikawa-Ankerhold
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; (M.K.); (S.B.); (P.G.); (H.I.-A.); (M.L.)
- Department of Internal Medicine I, Faculty of Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Klaus T. Preissner
- Department of Biochemistry, Faculty of Medicine, Justus Liebig University, 35392 Giessen, Germany; (L.L.P.); (K.T.P.); (S.F.)
| | - Silvia Fischer
- Department of Biochemistry, Faculty of Medicine, Justus Liebig University, 35392 Giessen, Germany; (L.L.P.); (K.T.P.); (S.F.)
| | - Manuel Lasch
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; (M.K.); (S.B.); (P.G.); (H.I.-A.); (M.L.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig- Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Elisabeth Deindl
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; (M.K.); (S.B.); (P.G.); (H.I.-A.); (M.L.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig- Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
- Correspondence: ; Tel.: +49-(0)-89-2180-76504
| |
Collapse
|
44
|
Aref Z, Quax PHA. In Vivo Matrigel Plug Assay as a Potent Method to Investigate Specific Individual Contribution of Angiogenesis to Blood Flow Recovery in Mice. Int J Mol Sci 2021; 22:ijms22168909. [PMID: 34445616 PMCID: PMC8396178 DOI: 10.3390/ijms22168909] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/26/2021] [Accepted: 08/17/2021] [Indexed: 12/24/2022] Open
Abstract
Neovascularization restores blood flow recovery after ischemia in peripheral arterial disease. The main two components of neovascularization are angiogenesis and arteriogenesis. Both of these processes contribute to functional improvements of blood flow after occlusion. However, discriminating between the specific contribution of each process is difficult. A frequently used model for investigating neovascularization is the murine hind limb ischemia model (HLI). With this model, it is difficult to determine the role of angiogenesis, because usually the timing for the sacrifice of the mice is chosen to be optimal for the analysis of arteriogenesis. More importantly, the occurring angiogenesis in the distal calf muscles is probably affected by the proximally occurring arteriogenesis. Therefore, to understand and subsequently intervene in the process of angiogenesis, a model is needed which investigates angiogenesis without the influence of arteriogenesis. In this study we evaluated the in vivo Matrigel plug assay in genetic deficient mice to investigate angiogenesis. Mice deficient for interferon regulatory factor (IRF)3, IRF7, RadioProtective 105 (RP105), Chemokine CC receptor CCR7, and p300/CBP-associated factor (PCAF) underwent the in vivo Matrigel model. Histological analysis of the Matrigel plugs showed an increased angiogenesis in mice deficient of IRF3, IRF7, and RP105, and a decreased angiogenesis in PCAF deficient mice. Our results also suggest an involvement of CCR7 in angiogenesis. Comparing our results with results of the HLI model found in the literature suggests that the in vivo Matrigel plug assay is superior in evaluating the angiogenic response after ischemia.
Collapse
Affiliation(s)
| | - Paul H. A. Quax
- Correspondence: ; Tel.: +31-71-526-1584; Fax: +31-71-526-6570
| |
Collapse
|
45
|
Abstract
The prevalence of peripheral arterial disease (PAD) in the United States exceeds 10 million people, and PAD is a significant cause of morbidity and mortality across the globe. PAD is typically caused by atherosclerotic obstructions in the large arteries to the leg(s). The most common clinical consequences of PAD include pain on walking (claudication), impaired functional capacity, pain at rest, and loss of tissue integrity in the distal limbs that may lead to lower extremity amputation. Patients with PAD also have higher than expected rates of myocardial infarction, stroke, and cardiovascular death. Despite advances in surgical and endovascular procedures, revascularization procedures may be suboptimal in relieving symptoms, and some patients with PAD cannot be treated because of comorbid conditions. In some cases, relieving obstructive disease in the large conduit arteries does not assure complete limb salvage because of severe microvascular disease. Despite several decades of investigational efforts, medical therapies to improve perfusion to the distal limb are of limited benefit. Whereas recent studies of anticoagulant (eg, rivaroxaban) and intensive lipid lowering (such as PCSK9 [proprotein convertase subtilisin/kexin type 9] inhibitors) have reduced major cardiovascular and limb events in PAD populations, chronic ischemia of the limb remains largely resistant to medical therapy. Experimental approaches to improve limb outcomes have included the administration of angiogenic cytokines (either as recombinant protein or as gene therapy) as well as cell therapy. Although early angiogenesis and cell therapy studies were promising, these studies lacked sufficient control groups and larger randomized clinical trials have yet to achieve significant benefit. This review will focus on what has been learned to advance medical revascularization for PAD and how that information might lead to novel approaches for therapeutic angiogenesis and arteriogenesis for PAD.
Collapse
Affiliation(s)
- Brian H Annex
- Vascular Biology Center, Department of Medicine, Medical College of Georgia, Augusta University (B.H.A.)
| | - John P Cooke
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, TX (J.P.C.)
| |
Collapse
|
46
|
Götz P, Braumandl A, Kübler M, Kumaraswami K, Ishikawa-Ankerhold H, Lasch M, Deindl E. C3 Deficiency Leads to Increased Angiogenesis and Elevated Pro-Angiogenic Leukocyte Recruitment in Ischemic Muscle Tissue. Int J Mol Sci 2021; 22:5800. [PMID: 34071589 PMCID: PMC8198161 DOI: 10.3390/ijms22115800] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 12/24/2022] Open
Abstract
The complement system is a potent inflammatory trigger, activator, and chemoattractant for leukocytes, which play a crucial role in promoting angiogenesis. However, little information is available about the influence of the complement system on angiogenesis in ischemic muscle tissue. To address this topic and analyze the impact of the complement system on angiogenesis, we induced muscle ischemia in complement factor C3 deficient (C3-/-) and wildtype control mice by femoral artery ligation (FAL). At 24 h and 7 days after FAL, we isolated the ischemic gastrocnemius muscles and investigated them by means of (immuno-)histological analyses. C3-/- mice showed elevated ischemic damage 7 days after FAL, as evidenced by H&E staining. In addition, angiogenesis was increased in C3-/- mice, as demonstrated by increased capillary/muscle fiber ratio and increased proliferating endothelial cells (CD31+/BrdU+). Moreover, our results showed that the total number of leukocytes (CD45+) was increased in C3-/- mice, which was based on an increased number of neutrophils (MPO+), neutrophil extracellular trap formation (MPO+/CitH3+), and macrophages (CD68+) displaying a shift toward an anti-inflammatory and pro-angiogenic M2-like polarized phenotype (CD68+/MRC1+). In summary, we show that the deficiency of complement factor C3 increased neutrophil and M2-like polarized macrophage accumulation in ischemic muscle tissue, contributing to angiogenesis.
Collapse
Affiliation(s)
- Philipp Götz
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; (P.G.); (A.B.); (M.K.); (K.K.); (H.I.-A.); (M.L.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Faculty of Medicine, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Anna Braumandl
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; (P.G.); (A.B.); (M.K.); (K.K.); (H.I.-A.); (M.L.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Faculty of Medicine, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Matthias Kübler
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; (P.G.); (A.B.); (M.K.); (K.K.); (H.I.-A.); (M.L.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Faculty of Medicine, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Konda Kumaraswami
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; (P.G.); (A.B.); (M.K.); (K.K.); (H.I.-A.); (M.L.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Faculty of Medicine, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Hellen Ishikawa-Ankerhold
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; (P.G.); (A.B.); (M.K.); (K.K.); (H.I.-A.); (M.L.)
- Department of Internal Medicine I, Faculty of Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Manuel Lasch
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; (P.G.); (A.B.); (M.K.); (K.K.); (H.I.-A.); (M.L.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Faculty of Medicine, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Elisabeth Deindl
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; (P.G.); (A.B.); (M.K.); (K.K.); (H.I.-A.); (M.L.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Faculty of Medicine, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| |
Collapse
|
47
|
Chen M, Li X. Role of TRPV4 channel in vasodilation and neovascularization. Microcirculation 2021; 28:e12703. [PMID: 33971061 DOI: 10.1111/micc.12703] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 05/02/2021] [Indexed: 12/12/2022]
Abstract
The transient receptor potential vanilloid type 4 (TRPV4) channel, a Ca2+ -permeable nonselective cation channel, is widely distributed in the circulatory system, particularly in vascular endothelial cells (ECs) and smooth muscle cells (SMCs). The TRPV4 channel is activated by various endogenous and exogenous stimuli, including shear stress, low intravascular pressure, and arachidonic acid. TRPV4 has a role in mediating vascular tone and arterial blood pressure. The activation of the TRPV4 channel induces Ca2+ influx, thereby resulting in endothelium-dependent hyperpolarization and SMC relaxation through SKCa and IKCa activation on ECs or through BKCa activation on SMCs. Ca2+ binds to calmodulin, which leads to the production of nitric oxide, causing vasodilation. Furthermore, the TRPV4 channel plays an important role in angiogenesis and arteriogenesis and is critical for tumor angiogenesis and growth, since it promotes or inhibits the development of various types of cancer. The TRPV4 channel is involved in the active growth of collateral arteries induced by flow shear stress, which makes it a promising therapeutic target in the occlusion or stenosis of the main arteries. In this review, we explore the role and the potential mechanism of action of the TRPV4 channel in the regulation of vascular tone and in the induction of neovascularization to provide a reference for future research.
Collapse
Affiliation(s)
- Miao Chen
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, Changchun, China
| | - Xiucun Li
- Department of Hand and Foot Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Anatomy and Histoembryology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
48
|
Hashim SNM, Yusof MFH, Zahari W, Noordin KBAA, Akamatsu T, Azlina A. Amniotic membrane matrix effects on calcineurin-NFAT-related gene expressions of SHED treated with VEGF for endothelial differentiation. In Vitro Cell Dev Biol Anim 2021; 57:560-570. [PMID: 34021476 DOI: 10.1007/s11626-021-00588-0] [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: 01/21/2021] [Accepted: 05/02/2021] [Indexed: 11/26/2022]
Abstract
The nuclear factor of activated T-cell (NFAT) signaling pathway is involved in angiogenesis following initiation by vascular endothelial growth factor (VEGF). A number of angiogenic genes have been associated with calcineurin in the NFAT pathway, forming a calcineurin-NFAT pathway. This study aims to investigate the involvement of four angiogenic genes within the calcineurin-NFAT pathway in the endothelial-like differentiation of stem cells from human exfoliated deciduous teeth (SHED) cultured on a human amniotic membrane (HAM) induced by VEGF. SHED were induced with VEGF for 24 h, then cultured on the stromal side of HAM. The cells were then further induced with VEGF until days 1 and 14. To understand the role of calcineurin, its potent inhibitor, cyclosporin A (CsA), was added into the culture. Results from SEM and H&E analyses showed SHED grew on HAM surface. Gene expression study of Cox-2 showed a drastically reduced expression with CsA treatment indicating Cox-2 involvement in the calcineurin-NFAT pathway. Meanwhile, IL-8 was probably controlled by another pathway as it showed no CsA inhibition. In contrast, high expression of ICAM-1 and RCAN1.4 by VEGF and CsA implied that these genes were not controlled by the calcineurin-NFAT-dependent pathway. In conclusion, the results of this study suggest the involvement of Cox-2 in the calcineurin-NFAT-dependent pathway while RCAN1.4 was controlled by NFAT molecule in endothelial-like differentiation of SHED cultured on HAM with VEGF induction.
Collapse
Affiliation(s)
- Siti Nurnasihah Md Hashim
- School of Dental Sciences, Health Campus, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Muhammad Fuad Hilmi Yusof
- School of Dental Sciences, Health Campus, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Wafa' Zahari
- School of Dental Sciences, Health Campus, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | | | - Tetsuya Akamatsu
- Department of Bioengineering, Division of Bioscience and Bioindustry, Graduate School of Technology, Industrial & Social Sciences, Tokushima University, Tokushima-shi, Tokushima, 770-8513, Japan
| | - Ahmad Azlina
- School of Dental Sciences, Health Campus, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia.
- Human Genome Centre, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia.
| |
Collapse
|
49
|
Makowski LM, Leffers M, Waltenberger J, Pardali E. Transforming growth factor-β1 signalling triggers vascular endothelial growth factor resistance and monocyte dysfunction in type 2 diabetes mellitus. J Cell Mol Med 2021; 25:5316-5325. [PMID: 33942489 PMCID: PMC8178271 DOI: 10.1111/jcmm.16543] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 03/07/2021] [Accepted: 03/24/2021] [Indexed: 12/01/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) leads to monocyte dysfunction associated with atherogenesis and defective arteriogenesis. Transforming growth factor (TGF)-β1, placenta growth factor (PlGF)-1 and vascular endothelial growth factor (VEGF)A play important roles in atherogenesis and arteriogenesis. VEGF-receptor (VEGFR)-mediated monocyte migration is inhibited in T2DM (VEGFA resistance), while TGF-β1-induced monocyte migration is fully functional. Therefore, we hypothesize that TGF-β antagonises the VEGFA responses in human monocytes. We demonstrate that monocytes from T2DM patients have an increased migratory response towards low concentrations of TGF-β1, while PlGF-1/VEGFA responses are mitigated. Mechanistically, this is due to increased expression of type II TGF-β receptor in monocytes under high-glucose conditions and increased expression of soluble (s)VEGFR1, which is known to interfere with VEGFA signalling. VEGFA resistance in monocytes from T2DM patients can be rescued by either experimental down-regulation of TGF-β receptor expression in vitro or by functional blocking of TGF-β signalling using either a TGF-β receptor kinase inhibitor or a TGF-β neutralizing antibody. Our data demonstrate that both T2DM and high-glucose potentiate the TGF-β pathway. TGF-β signalling impairs VEGFR-mediated responses in T2DM monocytes and in this way contributes to mononuclear cell dysfunction, provide novel insights into T2DM vascular dysfunction.
Collapse
Affiliation(s)
- Lena-Maria Makowski
- Department of Cardiology I-Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, Cardiolology, Münster, Germany
| | - Merle Leffers
- Department of Cardiology I-Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, Cardiolology, Münster, Germany
| | - Johannes Waltenberger
- Department of Cardiology I-Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, Cardiolology, Münster, Germany.,Cells-in-Motion Cluster of Excellence (EXC 1003-CiM), University of Münster, Münster, Germany.,Department of Cardiovascular Medicine, Medical Faculty, University of Münster, Münster, Germany
| | - Evangelia Pardali
- Department of Cardiology I-Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, Cardiolology, Münster, Germany.,Cells-in-Motion Cluster of Excellence (EXC 1003-CiM), University of Münster, Münster, Germany
| |
Collapse
|
50
|
Ding R, Yin YL, Jiang LH. Reactive Oxygen Species-Induced TRPM2-Mediated Ca 2+ Signalling in Endothelial Cells. Antioxidants (Basel) 2021; 10:antiox10050718. [PMID: 34063677 PMCID: PMC8147627 DOI: 10.3390/antiox10050718] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/21/2021] [Accepted: 04/28/2021] [Indexed: 02/07/2023] Open
Abstract
Endothelial cells form the innermost layer of blood vessels with a fundamental role as the physical barrier. While regulation of endothelial cell function by reactive oxygen species (ROS) is critical in physiological processes such as angiogenesis, endothelial function is a major target for interruption by oxidative stress resulting from generation of high levels of ROS in endothelial cells by various pathological factors and also release of ROS by neutrophils. TRPM2 is a ROS-sensitive Ca2+-permeable channel expressed in endothelial cells of various vascular beds. In this review, we provide an overview of the TRPM2 channel and its role in mediating ROS-induced Ca2+ signaling in endothelial cells. We discuss the TRPM2-mediated Ca2+ signaling in vascular endothelial growth factor-induced angiogenesis and in post-ischemic neovascularization. In particular, we examine the accumulative evidence that supports the role of TRPM2-mediated Ca2+ signaling in endothelial cell dysfunction caused by various oxidative stress-inducing factors that are associated with tissue inflammation, obesity and diabetes, as well as air pollution. These findings provide new, mechanistic insights into ROS-mediated regulation of endothelial cells in physiology and diseases.
Collapse
Affiliation(s)
- Ran Ding
- Department of Physiology and Pathophysiology, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Xinxiang Medical University, Xinxiang 453003, China; (R.D.); (Y.-L.Y.)
| | - Ya-Ling Yin
- Department of Physiology and Pathophysiology, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Xinxiang Medical University, Xinxiang 453003, China; (R.D.); (Y.-L.Y.)
| | - Lin-Hua Jiang
- Department of Physiology and Pathophysiology, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Xinxiang Medical University, Xinxiang 453003, China; (R.D.); (Y.-L.Y.)
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
- Correspondence: ; Tel.: +44-113-3434-231
| |
Collapse
|