1
|
Castillo-González J, González-Rey E. Beyond wrecking a wall: revisiting the concept of blood-brain barrier breakdown in ischemic stroke. Neural Regen Res 2025; 20:1944-1956. [PMID: 39254550 PMCID: PMC11691464 DOI: 10.4103/nrr.nrr-d-24-00392] [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: 04/10/2024] [Revised: 06/17/2024] [Accepted: 07/04/2024] [Indexed: 09/11/2024] Open
Abstract
The blood-brain barrier constitutes a dynamic and interactive boundary separating the central nervous system and the peripheral circulation. It tightly modulates the ion transport and nutrient influx, while restricting the entry of harmful factors, and selectively limiting the migration of immune cells, thereby maintaining brain homeostasis. Despite the well-established association between blood-brain barrier disruption and most neurodegenerative/neuroinflammatory diseases, much remains unknown about the factors influencing its physiology and the mechanisms underlying its breakdown. Moreover, the role of blood-brain barrier breakdown in the translational failure underlying therapies for brain disorders is just starting to be understood. This review aims to revisit this concept of "blood-brain barrier breakdown," delving into the most controversial aspects, prevalent challenges, and knowledge gaps concerning the lack of blood-brain barrier integrity. By moving beyond the oversimplistic dichotomy of an "open"/"bad" or a "closed"/"good" barrier, our objective is to provide a more comprehensive insight into blood-brain barrier dynamics, to identify novel targets and/or therapeutic approaches aimed at mitigating blood-brain barrier dysfunction. Furthermore, in this review, we advocate for considering the diverse time- and location-dependent alterations in the blood-brain barrier, which go beyond tight-junction disruption or brain endothelial cell breakdown, illustrated through the dynamics of ischemic stroke as a case study. Through this exploration, we seek to underscore the complexity of blood-brain barrier dysfunction and its implications for the pathogenesis and therapy of brain diseases.
Collapse
Affiliation(s)
- Julia Castillo-González
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, Granada, Spain
| | - Elena González-Rey
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, Granada, Spain
| |
Collapse
|
2
|
Brown TK, Dang C, Del Carmen A, Alharbi S, Chao CL, Xiong L, John NW, Smires A, Ho KJ, Jiang B. Mice Models for Peripheral Denervation to Enhance Vascular Regeneration. Tissue Eng Part C Methods 2025; 31:119-129. [PMID: 40062562 DOI: 10.1089/ten.tec.2025.0014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025] Open
Abstract
Sympathetic innervation plays a critical role in regulating vascular function, yet its influence on vascular regeneration and reinnervation following ischemic injury remains poorly understood. This study develops and validates murine models of localized sympathetic denervation using 6-hydroxydopamine (6-OHDA) to enable study of the sympathetic nervous system's impact on vascular systems during tissue repair. Two methods of 6-OHDA administration were employed: a single topical application during open surgery and minimally invasive weekly subcutaneous injections. The topical application model achieved temporary denervation lasting 1 week without causing vascular damage, while the subcutaneous injection model provided sustained denervation for up to 4 weeks with minimal inflammation and no significant changes to vascular architecture. To investigate the effects of denervation in an ischemic context, these models were combined with a hindlimb ischemia model. Ischemia induced persistent denervation in both 6-OHDA-treated and control limbs, with limited sympathetic nerve regeneration observed over 4 weeks. Despite persistent denervation, microvascular density and perfusion recovery in ischemic muscles were comparable between denervated and control groups. This suggests that ischemia governs vascular regeneration independently of sympathetic input. These results demonstrate that localized 6-OHDA administration provides a versatile tool for achieving controlled sympathetic denervation in peripheral arteries. These models provide a novel platform for studying vascular regeneration and reinnervation under both normal and ischemic conditions, offering novel insights into the interactions between neural regulation and vascular repair processes. This work lays the foundation for future research into neural-vascular crosstalk and new possibilities for developing regenerative therapies targeting the autonomic regulation of vascular health.
Collapse
Affiliation(s)
- Taylor K Brown
- Department of Biomedical Engineering, Northwestern University, Chicago, Illinois, USA
| | - Caitlyn Dang
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Aurea Del Carmen
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Sara Alharbi
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Calvin L Chao
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Liqun Xiong
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Nikita Wilson John
- Department of Biomedical Engineering, Northwestern University, Chicago, Illinois, USA
| | - Aidan Smires
- Department of Biomedical Engineering, Northwestern University, Chicago, Illinois, USA
| | - Karen J Ho
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Bin Jiang
- Department of Biomedical Engineering, Northwestern University, Chicago, Illinois, USA
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
3
|
Liberale L, Tual-Chalot S, Sedej S, Ministrini S, Georgiopoulos G, Grunewald M, Bäck M, Bochaton-Piallat ML, Boon RA, Ramos GC, de Winther MPJ, Drosatos K, Evans PC, Ferguson JF, Forslund-Startceva SK, Goettsch C, Giacca M, Haendeler J, Kallikourdis M, Ketelhuth DFJ, Koenen RR, Lacolley P, Lutgens E, Maffia P, Miwa S, Monaco C, Montecucco F, Norata GD, Osto E, Richardson GD, Riksen NP, Soehnlein O, Spyridopoulos I, Van Linthout S, Vilahur G, Wentzel JJ, Andrés V, Badimon L, Benetos A, Binder CJ, Brandes RP, Crea F, Furman D, Gorbunova V, Guzik TJ, Hill JA, Lüscher TF, Mittelbrunn M, Nencioni A, Netea MG, Passos JF, Stamatelopoulos KS, Tavernarakis N, Ungvari Z, Wu JC, Kirkland JL, Camici GG, Dimmeler S, Kroemer G, Abdellatif M, Stellos K. Roadmap for alleviating the manifestations of ageing in the cardiovascular system. Nat Rev Cardiol 2025:10.1038/s41569-025-01130-5. [PMID: 39972009 DOI: 10.1038/s41569-025-01130-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/22/2025] [Indexed: 02/21/2025]
Abstract
Ageing of the cardiovascular system is associated with frailty and various life-threatening diseases. As global populations grow older, age-related conditions increasingly determine healthspan and lifespan. The circulatory system not only supplies nutrients and oxygen to all tissues of the human body and removes by-products but also builds the largest interorgan communication network, thereby serving as a gatekeeper for healthy ageing. Therefore, elucidating organ-specific and cell-specific ageing mechanisms that compromise circulatory system functions could have the potential to prevent or ameliorate age-related cardiovascular diseases. In support of this concept, emerging evidence suggests that targeting the circulatory system might restore organ function. In this Roadmap, we delve into the organ-specific and cell-specific mechanisms that underlie ageing-related changes in the cardiovascular system. We raise unanswered questions regarding the optimal design of clinical trials, in which markers of biological ageing in humans could be assessed. We provide guidance for the development of gerotherapeutics, which will rely on the technological progress of the diagnostic toolbox to measure residual risk in elderly individuals. A major challenge in the quest to discover interventions that delay age-related conditions in humans is to identify molecular switches that can delay the onset of ageing changes. To overcome this roadblock, future clinical trials need to provide evidence that gerotherapeutics directly affect one or several hallmarks of ageing in such a manner as to delay, prevent, alleviate or treat age-associated dysfunction and diseases.
Collapse
Affiliation(s)
- Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Simon Tual-Chalot
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK.
| | - Simon Sedej
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | - Stefano Ministrini
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | | | - Myriam Grunewald
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Magnus Bäck
- Translational Cardiology, Centre for Molecular Medicine, Department of Medicine Solna, and Department of Cardiology, Heart and Vascular Centre, Karolinska Institutet, Stockholm, Sweden
- Inserm, DCAC, Université de Lorraine, Nancy, France
| | | | - Reinier A Boon
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC location VUmc, Amsterdam, Netherlands
| | - Gustavo Campos Ramos
- Department of Internal Medicine I/Comprehensive Heart Failure Centre, University Hospital Würzburg, Würzburg, Germany
| | - Menno P J de Winther
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences: Atherosclerosis and Ischaemic Syndromes; Amsterdam Infection and Immunity: Inflammatory Diseases, Amsterdam UMC location AMC, Amsterdam, Netherlands
| | - Konstantinos Drosatos
- Metabolic Biology Laboratory, Cardiovascular Center, Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Paul C Evans
- William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Jane F Ferguson
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sofia K Forslund-Startceva
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Claudia Goettsch
- Department of Internal Medicine I, Division of Cardiology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Mauro Giacca
- British Heart foundation Centre of Reseach Excellence, King's College London, London, UK
| | - Judith Haendeler
- Cardiovascular Degeneration, Medical Faculty, University Hospital and Heinrich-Heine University, Düsseldorf, Germany
| | - Marinos Kallikourdis
- Adaptive Immunity Lab, IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Daniel F J Ketelhuth
- Cardiovascular and Renal Research Unit, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Rory R Koenen
- CARIM-School for Cardiovascular Diseases, Department of Biochemistry, Maastricht University, Maastricht, Netherlands
| | | | - Esther Lutgens
- Department of Cardiovascular Medicine & Immunology, Mayo Clinic, Rochester, MN, USA
| | - Pasquale Maffia
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Satomi Miwa
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Claudia Monaco
- Kennedy Institute, NDORMS, University of Oxford, Oxford, UK
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Elena Osto
- Division of Physiology and Pathophysiology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria
| | - Gavin D Richardson
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Niels P Riksen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Oliver Soehnlein
- Institute of Experimental Pathology, University of Münster, Münster, Germany
| | - Ioakim Spyridopoulos
- Translational and Clinical Research Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Sophie Van Linthout
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätmedizin Berlin, Berlin, Germany
| | - Gemma Vilahur
- Research Institute, Hospital de la Santa Creu y Sant Pau l, IIB-Sant Pau, Barcelona, Spain
| | - Jolanda J Wentzel
- Cardiology, Biomedical Engineering, Erasmus MC, Rotterdam, Netherlands
| | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), CIBERCV, Madrid, Spain
| | - Lina Badimon
- Cardiovascular Health and Innovation Research Foundation (FICSI) and Cardiovascular Health and Network Medicine Department, University of Vic (UVIC-UCC), Barcelona, Spain
| | - Athanase Benetos
- Department of Geriatrics, University Hospital of Nancy and Inserm DCAC, Université de Lorraine, Nancy, France
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Ralf P Brandes
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt am Main, Germany
| | - Filippo Crea
- Centre of Excellence of Cardiovascular Sciences, Ospedale Isola Tiberina - Gemelli Isola, Roma, Italy
| | - David Furman
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Vera Gorbunova
- Departments of Biology and Medicine, University of Rochester, Rochester, NY, USA
| | - Tomasz J Guzik
- Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh, UK
| | - Joseph A Hill
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Thomas F Lüscher
- Heart Division, Royal Brompton and Harefield Hospital and National Heart and Lung Institute, Imperial College, London, UK
| | - María Mittelbrunn
- Consejo Superior de Investigaciones Científicas (CSIC), Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Alessio Nencioni
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
- Dipartimento di Medicina Interna e Specialità Mediche-DIMI, Università degli Studi di Genova, Genova, Italy
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - João F Passos
- Department of Physiology and Biomedical Engineering, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Kimon S Stamatelopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Nektarios Tavernarakis
- Medical School, University of Crete, and Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
| | - Zoltan Ungvari
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - James L Kirkland
- Center for Advanced Gerotherapeutics, Division of Endocrinology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Goethe University, Frankfurt am Main, Germany
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm, Institut Universitaire de France, Paris, France
| | | | - Konstantinos Stellos
- Department of Cardiovascular Research, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
4
|
Li W, Xu S, Chen L, Tan W, Deng N, Li Y, Zhang W, Deng C. Astragali Radix-Angelicae Sinensis Radix inhibits the activation of vascular adventitial fibroblasts and vascular intimal proliferation by regulating the TGF-β1/Smad2/3 pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119302. [PMID: 39743186 DOI: 10.1016/j.jep.2024.119302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 12/20/2024] [Accepted: 12/28/2024] [Indexed: 01/04/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Astragali Radix-Angelicae Sinensis Radix is an important traditional Chinese medicine used for the treatment of cardiovascular diseases. Our previous studies have shown that Astragali Radix-Angelicae Sinensis Radix can inhibit vascular intimal hyperplasia and improve the blood vessel wall's ECM deposition, among which six main active components can be absorbed into the blood, suggesting that these components may be the main pharmacodynamic substances of Astragali Radix-Angelicae Sinensis Radix against vascular intimal hyperplasia. AIM OF THE STUDY A mouse model of atherosclerosis was used to study the relationship between the anti-intimal hyperplasia effect of Astragali Radix-Angelicae Sinensis Radix and the inhibition of VAF activation and ECM synthesis. Furthermore, an in vitro rat VAF activation model was used. The effects of the main active ingredients of Astragali Radix-Angelicae Sinensis Radix on the proliferation, migration and ECM synthesis of VAF were observed. The mechanism of its action was investigated by focusing on TGF-β1/Smads signaling pathway. MATERIALS AND METHODS Male ApoE-/- mice were used to establish an AS model. Observe the morphological changes of blood vessels, the expression of Vimentin, α-SMA, ECM-related factors and TGF-β1/Smads signaling pathway-related proteins. Ang Ⅱ was used to induce the VAF activation model. The cell activity, cell proliferation, cell migration, cell phenotypic markers, ECM-related factors, cell cycle regulation-related proteins and TGF-β1/Smads signaling pathway-related proteins were determined. On this basis, TGF-β1/Smads signaling pathway agonists and inhibitors were used to study the effects of the compatibility of six active components on TGF-β1/Smads signaling pathway. RESULTS Astragali Radix-Angelicae Sinensis Radix can reduce aortic intimal hyperplasia, inhibit the expression of aortic α-SMA, Vimentin, ECM components, TGF-β1, p-Samd2 and p-Samd3. Cell experiments showed that the six active ingredients could inhibit the proliferation and migration of VAF to varying degrees, inhibit the expression of α-SMA, cell cycle promoters, ECM components, up-regulate the expression of Vimentin, P21, MMP2 and MMP9. The above effects were enhanced after the combination of the six components. The 6 components and their combinations could inhibit the expression of TGF-β1/Smads signaling pathway-related proteins and up-regulate the expression of Samd7 to varying degrees. The above effects were enhanced after the combination of the 6 components. TGF-β1/Smads signaling pathway inhibitor LY2157299 showed similar effects with the six components. The inhibitory effects of the six active ingredients on TGF-β1/Smads signaling pathway-related proteins and the promotion of Smad7 expression were attenuated when agonists were added into the six active ingredient combinations. However, adding TGFβ1/Smads signaling pathway inhibitor EGF to the six active ingredient combinations had no effect on the above effects. CONCLUSION Astragali Radix-Angelicae Sinensis Radix can inhibit intimal hyperplasia, VAF activation, and ECM synthesis in atherosclerosis. The six active ingredients may be the main pharmacological substances of Astragali Radix-Angelicae Sinensis Radix to inhibit the activation of VAF, and the combination of six ingredients can enhance their effects, which may be mediated by inhibiting the activation of the TGF-β1/Smad2/3 signaling pathway.
Collapse
Affiliation(s)
- Wanyu Li
- School of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, 300 Bachelor Road, Hanpu Science and Education Park, Yuelu District, 410208Changsha City, Hunan Province, China; Hunan Key Laboratory of Integrated Chinese and Western Medicine for Prevention and Treatment of Heart and Brain Diseases, 410208, Changsha, China.
| | - Shunzhou Xu
- The First Affiliated Hospital of Hunan Junior College of Traditional Chinese Medicine, No.571, Renmin Middle Road, Lusong District, 412008, Zhuzhou City, Hunan Province, China.
| | - Lingbo Chen
- Hunan Academy of Chinese Medicine, 142 Yuehua Road, Yuelu District, 410013, Changsha City, Hunan Province, China.
| | - Wei Tan
- School of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, 300 Bachelor Road, Hanpu Science and Education Park, Yuelu District, 410208Changsha City, Hunan Province, China; Hunan Key Laboratory of Integrated Chinese and Western Medicine for Prevention and Treatment of Heart and Brain Diseases, 410208, Changsha, China.
| | - Nujiao Deng
- The First Affiliated Hospital of Hunan University of Chinese Medicine, No. 95 Shaoshan Middle Road, Yuhua District, 410208, Changsha City, Hunan Province, China.
| | - Yanling Li
- School of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, 300 Bachelor Road, Hanpu Science and Education Park, Yuelu District, 410208Changsha City, Hunan Province, China; Hunan Key Laboratory of Integrated Chinese and Western Medicine for Prevention and Treatment of Heart and Brain Diseases, 410208, Changsha, China.
| | - Wei Zhang
- School of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, 300 Bachelor Road, Hanpu Science and Education Park, Yuelu District, 410208Changsha City, Hunan Province, China; Hunan Key Laboratory of Integrated Chinese and Western Medicine for Prevention and Treatment of Heart and Brain Diseases, 410208, Changsha, China.
| | - Changqing Deng
- School of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, 300 Bachelor Road, Hanpu Science and Education Park, Yuelu District, 410208Changsha City, Hunan Province, China; Hunan Key Laboratory of Integrated Chinese and Western Medicine for Prevention and Treatment of Heart and Brain Diseases, 410208, Changsha, China.
| |
Collapse
|
5
|
Yang H, Guo K, Ding P, Ning J, Zhang Y, Wang Y, Wang Z, Liu G, Shao C, Pan M, Ma Z, Yan X, Han J. Histone deacetylases: Regulation of vascular homeostasis via endothelial cells and vascular smooth muscle cells and the role in vascular pathogenesis. Genes Dis 2024; 11:101216. [PMID: 39281836 PMCID: PMC11396065 DOI: 10.1016/j.gendis.2024.101216] [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: 08/28/2023] [Revised: 12/20/2023] [Accepted: 01/04/2024] [Indexed: 09/18/2024] Open
Abstract
Histone deacetylases (HDACs) are proteases that play a key role in chromosome structural modification and gene expression regulation, and the involvement of HDACs in cancer, the nervous system, and the metabolic and immune system has been well reviewed. Our understanding of the function of HDACs in the vascular system has recently progressed, and a significant variety of HDAC inhibitors have been shown to be effective in the treatment of vascular diseases. However, few reviews have focused on the role of HDACs in the vascular system. In this study, the role of HDACs in the regulation of the vascular system mainly involving endothelial cells and vascular smooth muscle cells was discussed based on recent updates, and the role of HDACs in different vascular pathogenesis was summarized as well. Furthermore, the therapeutic effects and prospects of HDAC inhibitors were also addressed in this review.
Collapse
Affiliation(s)
- Hanyi Yang
- Department of Ophthalmology, Tangdu Hospital, The Air Force Military Medical University, Xi'an, Shaanxi 710038, China
- Xi'an Medical University, Xi'an, Shaanxi 710086, China
| | - Kai Guo
- Department of Thoracic Surgery, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710068, China
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, Shaanxi 710038, China
| | - Peng Ding
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, Shaanxi 710038, China
| | - Jiayi Ning
- Department of Ophthalmology, Tangdu Hospital, The Air Force Military Medical University, Xi'an, Shaanxi 710038, China
- Xi'an Medical University, Xi'an, Shaanxi 710086, China
| | - Yimeng Zhang
- Department of Ophthalmology, Tangdu Hospital, The Air Force Military Medical University, Xi'an, Shaanxi 710038, China
- Xi'an Medical University, Xi'an, Shaanxi 710086, China
| | - Yuanyong Wang
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, Shaanxi 710038, China
| | - Zhaoyang Wang
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, Shaanxi 710038, China
| | - Guanglin Liu
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, Shaanxi 710038, China
| | - Changjian Shao
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, Shaanxi 710038, China
| | - Minghong Pan
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, Shaanxi 710038, China
| | - Zhiqiang Ma
- Department of Medical Oncology, Senior Department of Oncology, Chinese PLA General Hospital, The Fifth Medical Center, Beijing 100853, China
| | - Xiaolong Yan
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, Shaanxi 710038, China
| | - Jing Han
- Department of Ophthalmology, Tangdu Hospital, The Air Force Military Medical University, Xi'an, Shaanxi 710038, China
| |
Collapse
|
6
|
Bi J, Cui D, Liu Z, Wang J, Chen Y, Wang S, Guo J, Dai X. Stent Graft-Induced High Wall Stress Promoted Aortic Wall Failure and Aortic Wall Injurious Complications After TEVAR: A Study of Numerical Simulation and Bioinformatics Analysis Based on Pig Models. J Endovasc Ther 2024:15266028241283324. [PMID: 39342458 DOI: 10.1177/15266028241283324] [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: 10/01/2024]
Abstract
OBJECTIVES Stent graft-related aortic injury is a major complication after thoracic endovascular aortic repair (TEVAR) and seriously affects patient prognosis. However, the distribution characteristics of aortic wall stress under the action of stent grafts and the mechanism of abnormal wall stress leading to aortic wall injury and adverse remodeling were unclear. The aim of this study was to explore the potential mechanisms of high wall stress on the structural and functional alterations of the aortic wall by combining animal experiments, numerical simulations, and bioinformatics. METHODS We observed stent graft-induced aortic injury by performing fenestrated TEVAR in 6 pigs, and quantitatively analyzed and visualized the stress distribution of the aortic wall under the stent graft through numerical simulation. Hematoxylin and eosin (HE) staining, Masson's trichrome staining, Verhoeff's Van Gieson (EVG) staining, and immunostaining were used to evaluate pathological changes in the aorta. Based on the numerical simulation results, the corresponding high-stress and low-stress regions of the aortic wall were subjected to bulk-RNA sequencing, and hub genes were identified by bioinformatics analysis. RESULTS Stent grafts were successfully implanted in 5 pigs. In all computational models, we found that obvious deformation and characteristic maximum stress concentration occurred on the side of the greater curve of the aortic arch in contact with the stent graft tip, and the high wall stress concentration areas were highly consistent with the obvious pathological injury area. Subsequent pathological analysis revealed that high wall stress-induced confusion and fragmentation of elastic fibers, collagen deposition, loss and phenotypic switching of vascular smooth muscle cells, and increased inflammatory responses. Gene expression profiles of the aortic wall under different wall stress conditions were described for the first time, and the hub genes (TGFB1, CDH5, DCN, ITGA5, ITGB3, and WT1) that may be involved in regulating the aortic injury and remodeling process in response to high wall stress stimulation were identified. CONCLUSIONS This study revealed a panoramic view of stent graft-associated high wall stress-induced aortic wall injury through technical approaches of multiple dimensions. Understanding these biomechanical features and hub genes is pivotal for advancing our comprehension of the complications associated with aortic injury after TEVAR and facilitating the development of future therapeutic interventions. CLINICAL IMPACT This study revealed a panoramic view of stent graft-associated high wall stress-induced aortic wall injury through technical approaches of multiple dimensions. The biomechanical distribution characteristics of the aortic wall, the secondary pathological injury and the alteration of gene expression profile under the action of stent graft were comprehensively revealed by animal experiments for the first time. This will advance clinicians' comprehension of complications associated with aortic injury after TEVAR, provide a new biomechanical perspective for the rational preoperative planning of TEVAR and the management of postoperative complications, and facilitate the development of future therapeutic interventions and stent graft device designs.
Collapse
Affiliation(s)
- Jiaxue Bi
- Department of Vascular Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin, China
| | - Dongsheng Cui
- Department of Vascular Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin, China
| | - Zongwei Liu
- Department of Vascular Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin, China
| | - Jiaxin Wang
- Department of Vascular Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin, China
| | - Yonghui Chen
- Department of Vascular Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin, China
| | - Shuaishuai Wang
- Department of Vascular Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin, China
| | - Jiayin Guo
- Department of Vascular Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin, China
| | - Xiangchen Dai
- Department of Vascular Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin, China
| |
Collapse
|
7
|
Veisman I, Massey WJ, Goren I, Liu W, Chauhan G, Rieder F. Muscular hyperplasia in Crohn's disease strictures: through thick and thin. Am J Physiol Cell Physiol 2024; 327:C671-C683. [PMID: 38912732 PMCID: PMC11427014 DOI: 10.1152/ajpcell.00307.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/18/2024] [Accepted: 06/18/2024] [Indexed: 06/25/2024]
Abstract
Fibrostenosing Crohn's disease (CD) represents a challenging clinical condition characterized by the development of symptomatic strictures within the gastrointestinal tract. Despite therapeutic advancements in managing inflammation, the progression of fibrostenotic complications remains a significant concern, often necessitating surgical intervention. Recent investigations have unveiled the pivotal role of smooth muscle cell hyperplasia in driving luminal narrowing and clinical symptomatology. Drawing parallels to analogous inflammatory conditions affecting other organs, such as the airways and blood vessels, sheds light on common underlying mechanisms of muscular hyperplasia. This review synthesizes current evidence to elucidate the mechanisms underlying smooth muscle cell proliferation in CD-associated strictures, offering insights into potential therapeutic targets. By highlighting the emerging significance of muscle thickening as a novel therapeutic target, this review aims to inform future research endeavors and clinical strategies with the goal to mitigate the burden of fibrostenotic complications in CD and other conditions.
Collapse
Affiliation(s)
- Ido Veisman
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States
| | - William J Massey
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States
| | - Idan Goren
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States
| | - Weiwei Liu
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States
| | - Gaurav Chauhan
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States
| | - Florian Rieder
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States
- Cleveland Clinic Program for Global Translational Inflammatory Bowel Diseases (GRID), Cleveland, Ohio, United States
| |
Collapse
|
8
|
Lu FT, Huang CC, Lai WY, Yang GY, Liang ZJ, Zhang ZY, Chokshi T, Guo KM, Tang YB, Chen Y, Yang ZH, Liang SJ, Pang RP, Zhou JG, Guan YY, Lv XF, Ma MM. Vascular smooth muscle-specific LRRC8A knockout ameliorates angiotensin II-induced cerebrovascular remodeling by inhibiting the WNK1/FOXO3a/MMP signaling pathway. Acta Pharmacol Sin 2024; 45:1848-1860. [PMID: 38719954 PMCID: PMC11335743 DOI: 10.1038/s41401-024-01280-1] [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: 11/28/2023] [Accepted: 03/27/2024] [Indexed: 08/22/2024]
Abstract
Hypertensive cerebrovascular remodeling involves the enlargement of vascular smooth muscle cells (VSMCs), which activates volume-regulated Cl- channels (VRCCs). The leucine-rich repeat-containing family 8 A (LRRC8A) has been shown to be the molecular identity of VRCCs. However, its role in vascular remodeling during hypertension is unclear. In this study, we used vascular smooth muscle-specific LRRC8A knockout (CKO) mice and an angiotensin II (Ang II)-induced hypertension model. The results showed that cerebrovascular remodeling during hypertension was ameliorated in CKO mice, and extracellular matrix (ECM) deposition was reduced. Based on the RNA-sequencing analysis of aortic tissues, the level of matrix metalloproteinases (MMPs), such as MMP-9 and MMP-14, were reduced in CKO mice with hypertension, which was further verified in vivo by qPCR and immunofluorescence analysis. Knockdown of LRRC8A in VSMCs inhibited the Ang II-induced upregulation of collagen I, fibronectin, and matrix metalloproteinases (MMPs), and overexpression of LRRC8A had the opposite effect. Further experiments revealed an interaction between with-no-lysine (K)-1 (WNK1), which is a "Cl--sensitive kinase", and Forkhead transcription factor O3a (FOXO3a), which is a transcription factor that regulates MMP expression. Ang II induced the phosphorylation of WNK1 and downstream FOXO3a, which then increased the expression of MMP-2 and MMP-9. This process was inhibited or potentiated when LRRC8A was knocked down or overexpressed, respectively. Overall, these results demonstrate that LRRC8A knockout in vascular smooth muscle protects against cerebrovascular remodeling during hypertension by reducing ECM deposition and inhibiting the WNK1/FOXO3a/MMP signaling pathway, demonstrating that LRRC8A is a potential therapeutic target for vascular remodeling-associated diseases such as stroke.
Collapse
Affiliation(s)
- Feng-Ting Lu
- Department of Pharmacology, and Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Department of Molecular Medicine, School of Medicine, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Cheng-Cui Huang
- Department of Pharmacy, the Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
| | - Wen-Yi Lai
- Department of Pharmacology, and Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Department of Molecular Medicine, School of Medicine, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Gui-Yong Yang
- Department of Pharmacology, and Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Zhu-Jun Liang
- Department of Pharmacology, and Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Zi-Yi Zhang
- Department of Pharmacology, and Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Tanvi Chokshi
- Research Division, Joslin Diabetes Center, Harvard University, Boston, MA, USA
| | - Kai-Min Guo
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Yu-Bo Tang
- Department of Pharmacy, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Yuan Chen
- Department of Molecular Medicine, School of Medicine, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Zhong-Han Yang
- Department of Molecular Medicine, School of Medicine, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Si-Jia Liang
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Rui-Ping Pang
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Jia-Guo Zhou
- Department of Pharmacology, and Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Yong-Yuan Guan
- Department of Pharmacology, and Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Xiao-Fei Lv
- Department of Pharmacology, and Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China.
| | - Ming-Ming Ma
- Department of Pharmacology, and Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China.
| |
Collapse
|
9
|
Zhang L, Feng Q, Kong W. ECM Microenvironment in Vascular Homeostasis: New Targets for Atherosclerosis. Physiology (Bethesda) 2024; 39:0. [PMID: 38984789 DOI: 10.1152/physiol.00028.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 03/05/2024] [Accepted: 03/23/2024] [Indexed: 07/11/2024] Open
Abstract
Alterations in vascular extracellular matrix (ECM) components, interactions, and mechanical properties influence both the formation and stability of atherosclerotic plaques. This review discusses the contribution of the ECM microenvironment in vascular homeostasis and remodeling in atherosclerosis, highlighting Cartilage oligomeric matrix protein (COMP) and its degrading enzyme ADAMTS7 as examples, and proposes potential avenues for future research aimed at identifying novel therapeutic targets for atherosclerosis based on the ECM microenvironment.
Collapse
Affiliation(s)
- Lu Zhang
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qianqian Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| |
Collapse
|
10
|
Liu L, Zhao B, Yu Y, Gao W, Liu W, Chen L, Xia Z, Cao Q. Vascular Aging in Ischemic Stroke. J Am Heart Assoc 2024; 13:e033341. [PMID: 39023057 PMCID: PMC11964078 DOI: 10.1161/jaha.123.033341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Cellular senescence, a permanent halt in cell division due to stress, spurs functional and structural changes, contributing to vascular aging characterized by endothelial dysfunction and vascular remodeling. This process raises the risk of ischemic stroke (IS) in older individuals, with its mechanisms still not completely understood despite ongoing research efforts. In this review, we have analyzed the impact of vascular aging on increasing susceptibility and exacerbating the pathology of IS. We have emphasized the detrimental effects of endothelial dysfunction and vascular remodeling influenced by oxidative stress and inflammatory response on vascular aging and IS. Our goal is to aid the understanding of vascular aging and IS pathogenesis, particularly benefiting older adults with high risk of IS.
Collapse
Affiliation(s)
- Lian Liu
- Department of AnesthesiologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Bo Zhao
- Department of AnesthesiologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Yueyang Yu
- Taikang Medical School, School of Basic Medical SciencesWuhan UniversityWuhanChina
| | - Wenwei Gao
- Department of Critical Care MedicineRenmin Hospital of Wuhan UniversityWuhanChina
| | - Weitu Liu
- Department of PathologyHubei Provincial Hospital of Traditional Chinese MedicineWuhanChina
| | - Lili Chen
- Department of AnesthesiologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Zhongyuan Xia
- Department of AnesthesiologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Quan Cao
- Department of NephrologyZhongnan Hospital of Wuhan UniversityWuhanChina
| |
Collapse
|
11
|
Pereira WDS, Lelis DF, Cunha RS, Griep RH, Barreto SM, Molina MDCB, Schmidt MI, Duncan BB, Bensenor I, Lotufo PA, Mill JG, Baldo MP. Fasting Glucose, Glycated Hemoglobin, and 2h Post-load Blood Glucose Are Independently Associated With Arterial Stiffness in Diabetes: The ELSA-Brasil Study. Angiology 2024; 75:635-644. [PMID: 36951393 DOI: 10.1177/00033197231166180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
The association of diabetes with increased large artery stiffness is not definitively established. We aimed to describe the carotid-femoral pulse wave velocity (cf-PWV) in participants with and without diabetes and whether the cf-PWV could vary among the different laboratory-based criteria used. A cross-sectional analysis using baseline data from 13,912 adults was used. cf-PWV as well as anthropometric, biochemical, and clinical data were measured. Diabetes was defined by previous medical diagnosis, medication use, fasting glucose, an oral glucose tolerance test (GTT), or glycated hemoglobin (HbA1c). The prevalence of diabetes was 18.7%, higher in men than in women. After adjustment, participants with diabetes showed higher cf-PWV (men: 9.7 ± 1.7 vs 9.4 ± 1.7 m/s, P < .05; women: 9.4 ± 1.6 vs 9.1 ± 1.7 m/s, P < .05). We observed a progressive increase in cf-PWV as >1 laboratory-based criterion for diabetes diagnosis was reached. Also, participants with diabetes with alterations in any laboratory-based criteria had higher cf-PWV than participants without diabetes, regardless of sex. In summary, diabetes is associated with higher cf-PWV as is each laboratory-based parameter used for its diagnosis. These results support the strong consequences of glucose dysregulation on the vascular system and provide evidence to screen all parameters involved in glycemic metabolism to improve vascular health.
Collapse
Affiliation(s)
- Wille D S Pereira
- Department of Pathophysiology, Montes Claros State University (UNIMONTES), Montes Claros, Brazil
| | - Deborah F Lelis
- Department of Pathophysiology, Montes Claros State University (UNIMONTES), Montes Claros, Brazil
| | - Roberto S Cunha
- Department of Physiological Sciences, Federal University of Espírito Santo, Vitória, Brazil
| | - Rosane H Griep
- Laboratory of Health and Environment Education, Oswaldo Cruz Institute, Rio de Janeiro, Brazil
| | - Sandhi M Barreto
- School of Medicine and Clinical Hospital/EBSERH, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Maria Inês Schmidt
- School of Medicine and Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Bruce B Duncan
- School of Medicine and Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Isabela Bensenor
- Center for Clinical and Epidemiologic Research, University of São Paulo, São Paulo, Brazil
| | - Paulo A Lotufo
- Center for Clinical and Epidemiologic Research, University of São Paulo, São Paulo, Brazil
| | - José Geraldo Mill
- Department of Physiological Sciences, Federal University of Espírito Santo, Vitória, Brazil
| | - Marcelo P Baldo
- Department of Pathophysiology, Montes Claros State University (UNIMONTES), Montes Claros, Brazil
| |
Collapse
|
12
|
Chandra Sekar N, Khoshmanesh K, Baratchi S. Bioengineered models of cardiovascular diseases. Atherosclerosis 2024; 393:117565. [PMID: 38714426 DOI: 10.1016/j.atherosclerosis.2024.117565] [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: 11/25/2023] [Revised: 04/15/2024] [Accepted: 04/25/2024] [Indexed: 05/09/2024]
Abstract
Age-associated cardiovascular diseases (CVDs), predominantly resulting from artery-related disorders such as atherosclerosis, stand as a leading cause of morbidity and mortality among the elderly population. Consequently, there is a growing interest in the development of clinically relevant bioengineered models of CVDs. Recent developments in bioengineering and material sciences have paved the way for the creation of intricate models that closely mimic the structure and surroundings of native cardiac tissues and blood vessels. These models can be utilized for basic research purposes and for identifying pharmaceutical interventions and facilitating drug discovery. The advancement of vessel-on-a-chip technologies and the development of bioengineered and humanized in vitro models of the cardiovascular system have the potential to revolutionize CVD disease modelling. These technologies offer pathophysiologically relevant models at a fraction of the cost and time required for traditional experimentation required in vivo. This progress signifies a significant advancement in the field, transitioning from conventional 2D cell culture models to advanced 3D organoid and vessel-on-a-chip models. These innovative models are specifically designed to explore the complexities of vascular aging and stiffening, crucial factors in the development of cardiovascular diseases. This review summarizes the recent progress of various bioengineered in vitro platforms developed for investigating the pathophysiology of human cardiovascular system with more focus on advanced 3D vascular platforms.
Collapse
Affiliation(s)
- Nadia Chandra Sekar
- School of Health & Biomedical Sciences, RMIT University, Bundoora, Victoria, 3082, Australia; Baker Heart and Diabetes Institute, Melbourne, Victoria, 3004, Australia
| | - Khashayar Khoshmanesh
- Baker Heart and Diabetes Institute, Melbourne, Victoria, 3004, Australia; School of Engineering, RMIT University, Melbourne, Victoria, 3000, Australia
| | - Sara Baratchi
- School of Health & Biomedical Sciences, RMIT University, Bundoora, Victoria, 3082, Australia; Baker Heart and Diabetes Institute, Melbourne, Victoria, 3004, Australia; Department of Cardiometabolic Health, The University of Melbourne, Parkville, Victoria, 3010, Australia.
| |
Collapse
|
13
|
Donahue CL, Badal RM, Younger TS, Guan W, Tolkacheva EG, Barocas VH. Atherosclerotic Calcifications Have a Local Effect on the Peel Behavior of Human Aortic Media. J Biomech Eng 2024; 146:061003. [PMID: 38329432 PMCID: PMC10983699 DOI: 10.1115/1.4064682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 02/01/2024] [Accepted: 02/02/2024] [Indexed: 02/09/2024]
Abstract
Aortic dissections, characterized by the propagation of a tear through the layers of the vessel wall, are critical, life-threatening events. Aortic calcifications are a common comorbidity in both acute and chronic dissections, yet their impact on dissection mechanics remains unclear. Using micro-computed tomography (CT) imaging, peel testing, and finite element modeling, this study examines the interplay between atherosclerotic calcifications and dissection mechanics. Samples cut from cadaveric human thoracic aortas were micro-CT imaged and subsequently peel-tested to map peel tension curves to the location of aortic calcifications. Empirical mode decomposition separated peel tension curves into high and low-frequency components, with high-frequency effects corresponding to interlamellar bonding mechanics and low-frequency effects to peel tension fluctuations. Finally, we used an idealized finite element model to examine how stiff calcifications affect aortic failure mechanics. Results showed that atherosclerosis influences dissection behavior on multiple length scales. Experimentally, atherosclerotic samples exhibited higher peel tensions and greater variance in the axial direction. The variation was driven by increased amplitudes of low-frequency tension fluctuations in diseased samples, indicating that more catastrophic propagations occur near calcifications. The simulations corroborated this finding, suggesting that the low-frequency changes resulted from the presence of a stiff calcification in the vessel wall. There were also modifications to the high-frequency peel mechanics, a response likely attributable to alterations in the microstructure and interlamellar bonding within the media. Considered collectively, these findings demonstrate that dissection mechanics are modified in aortic media nearby and adjacent to aortic calcifications.
Collapse
Affiliation(s)
- Carly L. Donahue
- Department of Biomedical Engineering, University of Minnesota, Nils Hasselmo Hall, Room 7-115, 321 Church St SE, Minneapolis, MN 55455
| | - Ruturaj M. Badal
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN 55455
| | - Thomas S. Younger
- Department of Biomedical Engineering, University of Minnesota, Nils Hasselmo Hall, Room 7-115, 321 Church St SE, Minneapolis, MN 55455
| | - Weihua Guan
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN 55455
| | - Elena G. Tolkacheva
- Department of Biomedical Engineering, University of Minnesota, Nils Hasselmo Hall, Room 7-115, 321 Church St SE, Minneapolis, MN 55455; Institute for Engineering in Medicine, University of Minnesota, Minneapolis, MN 55455
| | - Victor H. Barocas
- Department of Biomedical Engineering, University of Minnesota, Nils Hasselmo Hall, Room 7-115, 321 Church St SE, Minneapolis, MN 55455
| |
Collapse
|
14
|
Ventura-Antunes L, Nackenoff A, Romero-Fernandez W, Bosworth AM, Prusky A, Wang E, Carvajal-Tapia C, Shostak A, Harmsen H, Mobley B, Maldonado J, Solopova E, Caleb Snider J, David Merryman W, Lippmann ES, Schrag M. Arteriolar degeneration and stiffness in cerebral amyloid angiopathy are linked to β-amyloid deposition and lysyl oxidase. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.08.583563. [PMID: 38659767 PMCID: PMC11042178 DOI: 10.1101/2024.03.08.583563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Cerebral amyloid angiopathy (CAA) is a vasculopathy characterized by vascular β-amyloid (Aβ) deposition on cerebral blood vessels. CAA is closely linked to Alzheimer's disease (AD) and intracerebral hemorrhage. CAA is associated with the loss of autoregulation in the brain, vascular rupture, and cognitive decline. To assess morphological and molecular changes associated with the degeneration of penetrating arterioles in CAA, we analyzed post-mortem human brain tissue from 26 patients with mild, moderate, and severe CAA end neurological controls. The tissue was optically cleared for three-dimensional light sheet microscopy, and morphological features were quantified using surface volume rendering. We stained Aβ, vascular smooth muscle (VSM), lysyl oxidase (LOX), and vascular markers to visualize the relationship between degenerative morphological features, including vascular dilation, dolichoectasia (variability in lumenal diameter) and tortuosity, and the volumes of VSM, Aβ, and LOX in arterioles. Atomic force microscopy (AFM) was used to assess arteriolar wall stiffness, and we identified a pattern of morphological features associated with degenerating arterioles in the cortex. The volume of VSM associated with the arteriole was reduced by around 80% in arterioles with severe CAA and around 60% in cases with mild/moderate CAA. This loss of VSM correlated with increased arteriolar diameter and variability of diameter, suggesting VSM loss contributes to arteriolar laxity. These vascular morphological features correlated strongly with Aβ deposits. At sites of microhemorrhage, Aβ was consistently present, although the morphology of the deposits changed from the typical organized ring shape to sharply contoured shards with marked dilation of the vessel. AFM showed that arteriolar walls with CAA were more than 400% stiffer than those without CAA. Finally, we characterized the association of vascular degeneration with LOX, finding strong associations with VSM loss and vascular degeneration. These results show an association between vascular Aβ deposition, microvascular degeneration, and increased vascular stiffness, likely due to the combined effects of replacement of VSM by β-amyloid, cross-linking of extracellular matrices (ECM) by LOX, and possibly fibrosis. This advanced microscopic imaging study clarifies the association between Aβ deposition and vascular fragility. Restoration of physiologic ECM properties in penetrating arteries may yield a novel therapeutic strategy for CAA.
Collapse
Affiliation(s)
| | - Alex Nackenoff
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Allison M Bosworth
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Alex Prusky
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Emmeline Wang
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Alena Shostak
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hannah Harmsen
- Department of Pathology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bret Mobley
- Department of Pathology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jose Maldonado
- Vanderbilt Neurovisualization Lab, Vanderbilt University, Nashville, TN, USA
| | - Elena Solopova
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - J. Caleb Snider
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - W. David Merryman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Ethan S Lippmann
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville TN, USA
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Matthew Schrag
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville TN, USA
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
15
|
Fu Y, Zhou Y, Wang K, Li Z, Kong W. Extracellular Matrix Interactome in Modulating Vascular Homeostasis and Remodeling. Circ Res 2024; 134:931-949. [PMID: 38547250 DOI: 10.1161/circresaha.123.324055] [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] [Indexed: 04/02/2024]
Abstract
The ECM (extracellular matrix) is a major component of the vascular microenvironment that modulates vascular homeostasis. ECM proteins include collagens, elastin, noncollagen glycoproteins, and proteoglycans/glycosaminoglycans. ECM proteins form complex matrix structures, such as the basal lamina and collagen and elastin fibers, through direct interactions or lysyl oxidase-mediated cross-linking. Moreover, ECM proteins directly interact with cell surface receptors or extracellular secreted molecules, exerting matricellular and matricrine modulation, respectively. In addition, extracellular proteases degrade or cleave matrix proteins, thereby contributing to ECM turnover. These interactions constitute the ECM interactome network, which is essential for maintaining vascular homeostasis and preventing pathological vascular remodeling. The current review mainly focuses on endogenous matrix proteins in blood vessels and discusses the interaction of these matrix proteins with other ECM proteins, cell surface receptors, cytokines, complement and coagulation factors, and their potential roles in maintaining vascular homeostasis and preventing pathological remodeling.
Collapse
Affiliation(s)
- Yi Fu
- Department of Physiology and Pathophysiology (Y.F., K.W., Z.L., W.K.), School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yuan Zhou
- Department of Biomedical Informatics (Y.Z.), School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Kai Wang
- Department of Physiology and Pathophysiology (Y.F., K.W., Z.L., W.K.), School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Zhuofan Li
- Department of Physiology and Pathophysiology (Y.F., K.W., Z.L., W.K.), School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Wei Kong
- Department of Physiology and Pathophysiology (Y.F., K.W., Z.L., W.K.), School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| |
Collapse
|
16
|
Zhu L, Gou W, Ou L, Liu B, Liu M, Feng H. Role and new insights of microfibrillar-associated protein 4 in fibrotic diseases. APMIS 2024; 132:55-67. [PMID: 37957836 DOI: 10.1111/apm.13358] [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: 06/16/2023] [Accepted: 10/24/2023] [Indexed: 11/15/2023]
Abstract
Fibrosis is one of the most worrisome complications of chronic inflammatory diseases, leading to tissue damage, organ failure, and ultimately, death. The most notable pathological characteristic of fibrosis is the excessive accumulation of extracellular matrix (ECM) components such as collagen and fibronectin adjacent to foci of inflammation or damage. The human microfibrillar-associated protein 4 (MFAP4), an important member of the superfamily of fibrinogen-related proteins, is considered to have an extremely important role in ECM transformation of fibrogenesis. This review summarizes the structure, characteristics, and physiological functions of MFAP4 and the importance of MFAP4 in various fibrotic diseases. Meanwhile, we elaborated the underlying actions and mechanisms of MFAP4 in the development of fibrosis, suggesting that a better understand of MFAP4 broadens novel perspective for early screening, diagnosis, prognostic risk assessment, and treatment of fibrotic diseases.
Collapse
Affiliation(s)
- Long Zhu
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Changsha, China
- Xiangya Stomatological Hospital, Changsha, China
- Xiangya School of Stomatology, Central South University, Changsha, China
| | - Wenqun Gou
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Changsha, China
- Xiangya Stomatological Hospital, Changsha, China
- Xiangya School of Stomatology, Central South University, Changsha, China
- Changsha Stomatological Hospital, Changsha, China
| | - Lijia Ou
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Changsha, China
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Binjie Liu
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Changsha, China
- Xiangya Stomatological Hospital, Changsha, China
- Xiangya School of Stomatology, Central South University, Changsha, China
| | - Manyi Liu
- Xiangya Stomatological Hospital, Changsha, China
- Xiangya School of Stomatology, Central South University, Changsha, China
| | - Hui Feng
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Changsha, China
- Xiangya Stomatological Hospital, Changsha, China
- Xiangya School of Stomatology, Central South University, Changsha, China
| |
Collapse
|
17
|
Shabani Z, Schuerger J, Zhu X, Tang C, Ma L, Yadav A, Liang R, Press K, Weinsheimer S, Schmidt A, Wang C, Sekhar A, Nelson J, Kim H, Su H. Increased Collagen I/Collagen III Ratio Is Associated with Hemorrhage in Brain Arteriovenous Malformations in Human and Mouse. Cells 2024; 13:92. [PMID: 38201296 PMCID: PMC10778117 DOI: 10.3390/cells13010092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 12/23/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Background: The increase in the collagen I (COL I)/COL III ratio enhances vessel wall stiffness and renders vessels less resistant to blood flow and pressure changes. Activated microglia enhance inflammation-induced fibrosis. Hypotheses: The COL I/COL III ratio in human and mouse brain arteriovenous malformations (bAVMs) is associated with bAVM hemorrhage, and the depletion of microglia decreases the COL I/COL III ratio and hemorrhage. Method: COL I, COL III, and hemorrhages were analyzed in 12 human bAVMs and 6 control brains, and mouse bAVMs induced in three mouse lines with activin receptor-like kinase 1 (n = 7) or endoglin (n = 7) deleted in the endothelial cells or brain focally (n = 5). The controls for the mouse study were no-gene-deleted litter mates. Mouse bAVMs were used to test the relationships between the Col I/Col III ratio and hemorrhage and whether the transient depletion of microglia reduces the Col I/Col III ratio and hemorrhage. Results: The COL I/COL III ratio was higher in the human and mouse bAVMs than in controls. The microhemorrhage in mouse bAVMs was positively correlated with the Col I/Col III ratio. Transient depletion of microglia reduced the Col I/Col III ratio and microhemorrhage. Conclusions: The COL I/COL III ratio in the bAVMs was associated with bAVM hemorrhage. The depletion of microglia reduced the bAVM Col I/Col III ratio and hemorrhage.
Collapse
Affiliation(s)
- Zahra Shabani
- Center for Cerebrovascular Research, University of California, San Francisco, CA 94143, USA; (Z.S.); (J.S.); (X.Z.); (C.T.); (L.M.); (A.Y.); (R.L.); (K.P.); (S.W.); (A.S.); (C.W.); (A.S.); (J.N.); (H.K.)
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| | - Joana Schuerger
- Center for Cerebrovascular Research, University of California, San Francisco, CA 94143, USA; (Z.S.); (J.S.); (X.Z.); (C.T.); (L.M.); (A.Y.); (R.L.); (K.P.); (S.W.); (A.S.); (C.W.); (A.S.); (J.N.); (H.K.)
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| | - Xiaonan Zhu
- Center for Cerebrovascular Research, University of California, San Francisco, CA 94143, USA; (Z.S.); (J.S.); (X.Z.); (C.T.); (L.M.); (A.Y.); (R.L.); (K.P.); (S.W.); (A.S.); (C.W.); (A.S.); (J.N.); (H.K.)
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| | - Chaoliang Tang
- Center for Cerebrovascular Research, University of California, San Francisco, CA 94143, USA; (Z.S.); (J.S.); (X.Z.); (C.T.); (L.M.); (A.Y.); (R.L.); (K.P.); (S.W.); (A.S.); (C.W.); (A.S.); (J.N.); (H.K.)
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| | - Li Ma
- Center for Cerebrovascular Research, University of California, San Francisco, CA 94143, USA; (Z.S.); (J.S.); (X.Z.); (C.T.); (L.M.); (A.Y.); (R.L.); (K.P.); (S.W.); (A.S.); (C.W.); (A.S.); (J.N.); (H.K.)
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| | - Alka Yadav
- Center for Cerebrovascular Research, University of California, San Francisco, CA 94143, USA; (Z.S.); (J.S.); (X.Z.); (C.T.); (L.M.); (A.Y.); (R.L.); (K.P.); (S.W.); (A.S.); (C.W.); (A.S.); (J.N.); (H.K.)
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| | - Rich Liang
- Center for Cerebrovascular Research, University of California, San Francisco, CA 94143, USA; (Z.S.); (J.S.); (X.Z.); (C.T.); (L.M.); (A.Y.); (R.L.); (K.P.); (S.W.); (A.S.); (C.W.); (A.S.); (J.N.); (H.K.)
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| | - Kelly Press
- Center for Cerebrovascular Research, University of California, San Francisco, CA 94143, USA; (Z.S.); (J.S.); (X.Z.); (C.T.); (L.M.); (A.Y.); (R.L.); (K.P.); (S.W.); (A.S.); (C.W.); (A.S.); (J.N.); (H.K.)
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| | - Shantel Weinsheimer
- Center for Cerebrovascular Research, University of California, San Francisco, CA 94143, USA; (Z.S.); (J.S.); (X.Z.); (C.T.); (L.M.); (A.Y.); (R.L.); (K.P.); (S.W.); (A.S.); (C.W.); (A.S.); (J.N.); (H.K.)
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| | - Annika Schmidt
- Center for Cerebrovascular Research, University of California, San Francisco, CA 94143, USA; (Z.S.); (J.S.); (X.Z.); (C.T.); (L.M.); (A.Y.); (R.L.); (K.P.); (S.W.); (A.S.); (C.W.); (A.S.); (J.N.); (H.K.)
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| | - Calvin Wang
- Center for Cerebrovascular Research, University of California, San Francisco, CA 94143, USA; (Z.S.); (J.S.); (X.Z.); (C.T.); (L.M.); (A.Y.); (R.L.); (K.P.); (S.W.); (A.S.); (C.W.); (A.S.); (J.N.); (H.K.)
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| | - Abinav Sekhar
- Center for Cerebrovascular Research, University of California, San Francisco, CA 94143, USA; (Z.S.); (J.S.); (X.Z.); (C.T.); (L.M.); (A.Y.); (R.L.); (K.P.); (S.W.); (A.S.); (C.W.); (A.S.); (J.N.); (H.K.)
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| | - Jeffrey Nelson
- Center for Cerebrovascular Research, University of California, San Francisco, CA 94143, USA; (Z.S.); (J.S.); (X.Z.); (C.T.); (L.M.); (A.Y.); (R.L.); (K.P.); (S.W.); (A.S.); (C.W.); (A.S.); (J.N.); (H.K.)
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| | - Helen Kim
- Center for Cerebrovascular Research, University of California, San Francisco, CA 94143, USA; (Z.S.); (J.S.); (X.Z.); (C.T.); (L.M.); (A.Y.); (R.L.); (K.P.); (S.W.); (A.S.); (C.W.); (A.S.); (J.N.); (H.K.)
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| | - Hua Su
- Center for Cerebrovascular Research, University of California, San Francisco, CA 94143, USA; (Z.S.); (J.S.); (X.Z.); (C.T.); (L.M.); (A.Y.); (R.L.); (K.P.); (S.W.); (A.S.); (C.W.); (A.S.); (J.N.); (H.K.)
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
18
|
Hong IS. Endometrial Stem Cells: Orchestrating Dynamic Regeneration of Endometrium and Their Implications in Diverse Endometrial Disorders. Int J Biol Sci 2024; 20:864-879. [PMID: 38250149 PMCID: PMC10797688 DOI: 10.7150/ijbs.89795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 12/24/2023] [Indexed: 01/23/2024] Open
Abstract
The human endometrium, a vital component of the uterus, undergoes dynamic changes during the menstrual cycle to create a receptive environment for embryo implantation. Its remarkable regenerative capacity can be attributed to the presence of tissue-resident stem cell populations within the endometrium. Despite variations in characteristics among different subtypes, endometrial stem cells exhibit notably robust self-renewal capacity and the ability to differentiate into multiple lineages. This review offers a comprehensive insight into the current literature and recent advancements regarding the roles of various endometrial stem cell types during dynamic regeneration of the endometrium during the menstrual cycle. In addition, emerging evidence suggests that dysfunction or depletion of endometrial stem cells may play critical roles in the development and progression of various endometrial disorders, such as endometriosis, uterine fibroids, adenomyosis, infertility, and endometrial cancer. Therefore, we also highlight potential roles of endometrial stem cells in the development and progression of these endometrial diseases, including their ability to accumulate genetic mutations and express genes associated with endometrial diseases. Understanding the dynamic properties of the endometrium and the roles of endometrial stem cells in various endometrial disorders will shed light on potential therapeutic strategies for managing these conditions and improving women's fertility outcomes.
Collapse
Affiliation(s)
- In-Sun Hong
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, Republic of Korea
- Department of Molecular Medicine, School of Medicine, Gachon University, Incheon 406-840, Republic of Korea
| |
Collapse
|
19
|
Obaha A, Novinec M. Regulation of Peptidase Activity beyond the Active Site in Human Health and Disease. Int J Mol Sci 2023; 24:17120. [PMID: 38069440 PMCID: PMC10707025 DOI: 10.3390/ijms242317120] [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: 11/17/2023] [Revised: 12/01/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023] Open
Abstract
This comprehensive review addresses the intricate and multifaceted regulation of peptidase activity in human health and disease, providing a comprehensive investigation that extends well beyond the boundaries of the active site. Our review focuses on multiple mechanisms and highlights the important role of exosites, allosteric sites, and processes involved in zymogen activation. These mechanisms play a central role in shaping the complex world of peptidase function and are promising potential targets for the development of innovative drugs and therapeutic interventions. The review also briefly discusses the influence of glycosaminoglycans and non-inhibitory binding proteins on enzyme activities. Understanding their role may be a crucial factor in the development of therapeutic strategies. By elucidating the intricate web of regulatory mechanisms that control peptidase activity, this review deepens our understanding in this field and provides a roadmap for various strategies to influence and modulate peptidase activity.
Collapse
Affiliation(s)
| | - Marko Novinec
- Faculty of Chemistry and Chemical Technology, Department of Chemistry and Biochemistry, University of Ljubljana, Večna pot 113, SI-1000 Ljubljana, Slovenia;
| |
Collapse
|
20
|
Krajnik A, Nimmer E, Brazzo JA, Biber JC, Drewes R, Tumenbayar BI, Sullivan A, Pham K, Krug A, Heo Y, Kolega J, Heo SJ, Lee K, Weil BR, Kim DH, Gupte SA, Bae Y. Survivin regulates intracellular stiffness and extracellular matrix production in vascular smooth muscle cells. APL Bioeng 2023; 7:046104. [PMID: 37868708 PMCID: PMC10590228 DOI: 10.1063/5.0157549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 10/05/2023] [Indexed: 10/24/2023] Open
Abstract
Vascular dysfunction is a common cause of cardiovascular diseases characterized by the narrowing and stiffening of arteries, such as atherosclerosis, restenosis, and hypertension. Arterial narrowing results from the aberrant proliferation of vascular smooth muscle cells (VSMCs) and their increased synthesis and deposition of extracellular matrix (ECM) proteins. These, in turn, are modulated by arterial stiffness, but the mechanism for this is not fully understood. We found that survivin is an important regulator of stiffness-mediated ECM synthesis and intracellular stiffness in VSMCs. Whole-transcriptome analysis and cell culture experiments showed that survivin expression is upregulated in injured femoral arteries in mice and in human VSMCs cultured on stiff fibronectin-coated hydrogels. Suppressed expression of survivin in human VSMCs significantly decreased the stiffness-mediated expression of ECM components related to arterial stiffening, such as collagen-I, fibronectin, and lysyl oxidase. By contrast, expression of these ECM proteins was rescued by ectopic expression of survivin in human VSMCs cultured on soft hydrogels. Interestingly, atomic force microscopy analysis showed that suppressed or ectopic expression of survivin decreases or increases intracellular stiffness, respectively. Furthermore, we observed that inhibiting Rac and Rho reduces survivin expression, elucidating a mechanical pathway connecting intracellular tension, mediated by Rac and Rho, to survivin induction. Finally, we found that survivin inhibition decreases FAK phosphorylation, indicating that survivin-dependent intracellular tension feeds back to maintain signaling through FAK. These findings suggest a novel mechanism by which survivin potentially modulates arterial stiffness.
Collapse
Affiliation(s)
- Amanda Krajnik
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14203, USA
| | - Erik Nimmer
- Department of Biomedical Engineering, School of Engineering and Applied Sciences, University at Buffalo, Buffalo, New York 14260, USA
| | - Joseph A. Brazzo
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14203, USA
| | - John C. Biber
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14203, USA
| | - Rhonda Drewes
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14203, USA
| | - Bat-Ider Tumenbayar
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14203, USA
| | - Andra Sullivan
- Department of Biomedical Engineering, School of Engineering and Applied Sciences, University at Buffalo, Buffalo, New York 14260, USA
| | - Khanh Pham
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14203, USA
| | - Alanna Krug
- Department of Biomedical Engineering, School of Engineering and Applied Sciences, University at Buffalo, Buffalo, New York 14260, USA
| | | | - John Kolega
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14203, USA
| | - Su-Jin Heo
- Department of Orthopedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | - Brian R. Weil
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14203, USA
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21205, USA
| | - Sachin A. Gupte
- Department of Pharmacology, New York Medical College, Valhalla, New York 10595, USA
| | - Yongho Bae
- Author to whom correspondence should be addressed:
| |
Collapse
|
21
|
Yong J, Wang R, Song F, Wang T. The protective effects of pirfenidone in preventing abdominal aortic aneurysm formation. J Biochem Mol Toxicol 2023; 37:e23514. [PMID: 37691532 DOI: 10.1002/jbt.23514] [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: 10/09/2022] [Revised: 07/07/2023] [Accepted: 08/17/2023] [Indexed: 09/12/2023]
Abstract
Vascular endothelial growth factor (VEGF)-mediated angiogenesis participates in the initiation and progression of abdominal aortic aneurysm (AAA). Pirfenidone is a compound that has anti-inflammatory and antioxidant properties and suppresses angiogenesis. Pirfenidone targets the extracellular matrix (ECM) and has therapeutic effects on fibrotic diseases. Therefore, we speculated that pirfenidone might have meaningful therapeutic effects in AAA, and the current study was designed to investigate this capacity. An AAA model was constructed in mice using a long-term injection of angiotensin II (Ang II), followed by a 28-day administration of 200 mg/kg/day pirfenidone. Increased maximal external diameter of the abdominal artery, promoted levels of VEGF-A and its receptor VEGF-R2, upregulated matrix metallopeptidases (MMP)-2 and MMP-9, and elevated release of pro-inflammatory cytokines were observed in AAA mice, which were extremely repressed by 200 mg/kg pirfenidone. Human aortic endothelial cells (HAECs) were stimulated with Ang II for 1 day, in the presence or absence of pirfenidone (100 nM). Elevated expression of VEGF-A and VEGF-R2, facilitated proliferation, increased tube formation ability, and upregulated MMP-2 and MMP-9 were observed in Ang II-stimulated HAECs, all of which were significantly rescued by 100 nM pirfenidone. Finally, the elevated levels of myeloid differentiation primary response 88 and phosphorylated nuclear factor-kappa-B subunit p65 observed in Ang II-stimulated HAECs were repressed by pirfenidone. Collectively, pirfenidone alleviated AAA by inhibiting ECM degradation and ameliorating endothelial dysfunction.
Collapse
Affiliation(s)
- Jun Yong
- Department of Vascular Surgery, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Rui Wang
- Department of Vascular Surgery, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Fubo Song
- Department of Medical Records Room, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Tao Wang
- Department of Vascular Surgery, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| |
Collapse
|
22
|
Katoh K. Effects of Mechanical Stress on Endothelial Cells In Situ and In Vitro. Int J Mol Sci 2023; 24:16518. [PMID: 38003708 PMCID: PMC10671803 DOI: 10.3390/ijms242216518] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 11/26/2023] Open
Abstract
Endothelial cells lining blood vessels are essential for maintaining vascular homeostasis and mediate several pathological and physiological processes. Mechanical stresses generated by blood flow and other biomechanical factors significantly affect endothelial cell activity. Here, we review how mechanical stresses, both in situ and in vitro, affect endothelial cells. We review the basic principles underlying the cellular response to mechanical stresses. We also consider the implications of these findings for understanding the mechanisms of mechanotransducer and mechano-signal transduction systems by cytoskeletal components.
Collapse
Affiliation(s)
- Kazuo Katoh
- Laboratory of Human Anatomy and Cell Biology, Faculty of Health Sciences, Tsukuba University of Technology, Tsukuba 305-8521, Japan
| |
Collapse
|
23
|
Latifi-Navid H, Barzegar Behrooz A, Jamehdor S, Davari M, Latifinavid M, Zolfaghari N, Piroozmand S, Taghizadeh S, Bourbour M, Shemshaki G, Latifi-Navid S, Arab SS, Soheili ZS, Ahmadieh H, Sheibani N. Construction of an Exudative Age-Related Macular Degeneration Diagnostic and Therapeutic Molecular Network Using Multi-Layer Network Analysis, a Fuzzy Logic Model, and Deep Learning Techniques: Are Retinal and Brain Neurodegenerative Disorders Related? Pharmaceuticals (Basel) 2023; 16:1555. [PMID: 38004422 PMCID: PMC10674956 DOI: 10.3390/ph16111555] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/26/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
Neovascular age-related macular degeneration (nAMD) is a leading cause of irreversible visual impairment in the elderly. The current management of nAMD is limited and involves regular intravitreal administration of anti-vascular endothelial growth factor (anti-VEGF). However, the effectiveness of these treatments is limited by overlapping and compensatory pathways leading to unresponsiveness to anti-VEGF treatments in a significant portion of nAMD patients. Therefore, a system view of pathways involved in pathophysiology of nAMD will have significant clinical value. The aim of this study was to identify proteins, miRNAs, long non-coding RNAs (lncRNAs), various metabolites, and single-nucleotide polymorphisms (SNPs) with a significant role in the pathogenesis of nAMD. To accomplish this goal, we conducted a multi-layer network analysis, which identified 30 key genes, six miRNAs, and four lncRNAs. We also found three key metabolites that are common with AMD, Alzheimer's disease (AD) and schizophrenia. Moreover, we identified nine key SNPs and their related genes that are common among AMD, AD, schizophrenia, multiple sclerosis (MS), and Parkinson's disease (PD). Thus, our findings suggest that there exists a connection between nAMD and the aforementioned neurodegenerative disorders. In addition, our study also demonstrates the effectiveness of using artificial intelligence, specifically the LSTM network, a fuzzy logic model, and genetic algorithms, to identify important metabolites in complex metabolic pathways to open new avenues for the design and/or repurposing of drugs for nAMD treatment.
Collapse
Affiliation(s)
- Hamid Latifi-Navid
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran 1497716316, Iran; (H.L.-N.); (M.D.); (N.Z.); (S.P.); (S.T.); (Z.-S.S.)
- Departments of Ophthalmology and Visual Sciences and Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Amir Barzegar Behrooz
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3T 2N2, Canada;
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran 1416634793, Iran
| | - Saleh Jamehdor
- Department of Virology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan 6517838636, Iran;
| | - Maliheh Davari
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran 1497716316, Iran; (H.L.-N.); (M.D.); (N.Z.); (S.P.); (S.T.); (Z.-S.S.)
| | - Masoud Latifinavid
- Department of Mechatronic Engineering, University of Turkish Aeronautical Association, 06790 Ankara, Turkey;
| | - Narges Zolfaghari
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran 1497716316, Iran; (H.L.-N.); (M.D.); (N.Z.); (S.P.); (S.T.); (Z.-S.S.)
| | - Somayeh Piroozmand
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran 1497716316, Iran; (H.L.-N.); (M.D.); (N.Z.); (S.P.); (S.T.); (Z.-S.S.)
| | - Sepideh Taghizadeh
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran 1497716316, Iran; (H.L.-N.); (M.D.); (N.Z.); (S.P.); (S.T.); (Z.-S.S.)
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Mahsa Bourbour
- Department of Biotechnology, Alzahra University, Tehran 1993893973, Iran;
| | - Golnaz Shemshaki
- Department of Studies in Zoology, University of Mysore, Manasagangothri, Mysore 570005, India;
| | - Saeid Latifi-Navid
- Department of Biology, Faculty of Sciences, University of Mohaghegh Ardabili, Ardabil 5619911367, Iran;
| | - Seyed Shahriar Arab
- Biophysics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 1411713116, Iran;
| | - Zahra-Soheila Soheili
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran 1497716316, Iran; (H.L.-N.); (M.D.); (N.Z.); (S.P.); (S.T.); (Z.-S.S.)
| | - Hamid Ahmadieh
- Ophthalmic Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran 1666673111, Iran;
| | - Nader Sheibani
- Departments of Ophthalmology and Visual Sciences and Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| |
Collapse
|
24
|
Castillo-González J, Ruiz JL, Serrano-Martínez I, Forte-Lago I, Ubago-Rodriguez A, Caro M, Pérez-Gómez JM, Benítez-Troncoso A, Andrés-León E, Sánchez-Navarro M, Luque RM, González-Rey E. Cortistatin deficiency reveals a dysfunctional brain endothelium with impaired gene pathways, exacerbated immune activation, and disrupted barrier integrity. J Neuroinflammation 2023; 20:226. [PMID: 37794493 PMCID: PMC10548650 DOI: 10.1186/s12974-023-02908-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 09/26/2023] [Indexed: 10/06/2023] Open
Abstract
BACKGROUND Brain activity governing cognition and behaviour depends on the fine-tuned microenvironment provided by a tightly controlled blood-brain barrier (BBB). Brain endothelium dysfunction is a hallmark of BBB breakdown in most neurodegenerative/neuroinflammatory disorders. Therefore, the identification of new endogenous molecules involved in endothelial cell disruption is essential to better understand BBB dynamics. Cortistatin is a neuroimmune mediator with anti-inflammatory and neuroprotective properties that exerts beneficial effects on the peripheral endothelium. However, its role in the healthy and injured brain endothelium remains to be evaluated. Herein, this study aimed to investigate the potential function of endogenous and therapeutic cortistatin in regulating brain endothelium dysfunction in a neuroinflammatory/neurodegenerative environment. METHODS Wild-type and cortistatin-deficient murine brain endothelium and human cells were used for an in vitro barrier model, where a simulated ischemia-like environment was mimicked. Endothelial permeability, junction integrity, and immune response in the presence and absence of cortistatin were evaluated using different size tracers, immunofluorescence labelling, qPCR, and ELISA. Cortistatin molecular mechanisms underlying brain endothelium dynamics were assessed by RNA-sequencing analysis. Cortistatin role in BBB leakage was evaluated in adult mice injected with LPS. RESULTS The endogenous lack of cortistatin predisposes endothelium weakening with increased permeability, tight-junctions breakdown, and dysregulated immune activity. We demonstrated that both damaged and uninjured brain endothelial cells isolated from cortistatin-deficient mice, present a dysregulated and/or deactivated genetic programming. These pathways, related to basic physiology but also crucial for the repair after damage (e.g., extracellular matrix remodelling, angiogenesis, response to oxygen, signalling, and metabolites transport), are dysfunctional and make brain endothelial barrier lacking cortistatin non-responsive to any further injury. Treatment with cortistatin reversed in vitro hyperpermeability, tight-junctions disruption, inflammatory response, and reduced in vivo BBB leakage. CONCLUSIONS The neuropeptide cortistatin has a key role in the physiology of the cerebral microvasculature and its presence is crucial to develop a canonical balanced response to damage. The reparative effects of cortistatin in the brain endothelium were accompanied by the modulation of the immune function and the rescue of barrier integrity. Cortistatin-based therapies could emerge as a novel pleiotropic strategy to ameliorate neuroinflammatory/neurodegenerative disorders with disrupted BBB.
Collapse
Affiliation(s)
- Julia Castillo-González
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016, Granada, Spain
| | - José Luis Ruiz
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016, Granada, Spain
| | - Ignacio Serrano-Martínez
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016, Granada, Spain
| | - Irene Forte-Lago
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016, Granada, Spain
| | - Ana Ubago-Rodriguez
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016, Granada, Spain
| | - Marta Caro
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016, Granada, Spain
| | - Jesús Miguel Pérez-Gómez
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Cordoba, 14004, Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004, Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004, Cordoba, Spain
| | | | - Eduardo Andrés-León
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016, Granada, Spain
| | - Macarena Sánchez-Navarro
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016, Granada, Spain
| | - Raúl M Luque
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Cordoba, 14004, Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004, Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004, Cordoba, Spain
| | - Elena González-Rey
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016, Granada, Spain.
| |
Collapse
|
25
|
Zhang W, Lin W, Zeng X, Zhang M, Chen Q, Tang Y, Sun J, Liang B, Zha L, Yu Z. FUT8-Mediated Core Fucosylation Promotes the Pulmonary Vascular Remodeling in Pulmonary Arterial Hypertension. Aging Dis 2023; 14:1927-1944. [PMID: 37196106 PMCID: PMC10529761 DOI: 10.14336/ad.2023.0218] [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: 11/12/2022] [Accepted: 02/18/2023] [Indexed: 05/19/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive cardiopulmonary disease with unclear underlying molecular mechanisms and limited therapeutic options. This study aimed to explore the role of core fucosylation and the only glycosyltransferase FUT8 in PAH. We observed increased core fucosylation in a monocrotaline (MCT)-induced PAH rat model and isolated rat pulmonary artery smooth muscle cells (PASMCs) treated with platelet-derived growth factor-BB (PDGF-BB). We found that 2-fluorofucose (2FF), a drug used to inhibit core fucosylation, improved hemodynamics and pulmonary vascular remodeling in MCT-induced PAH rats. In vitro, 2FF effectively restrains the proliferation, migration, and phenotypic switching of PASMCs and promotes apoptosis. Compared with controls, serum FUT8 concentration in PAH patients and MCT-induced rats was significantly elevated. FUT8 expression appeared increased in the lung tissues of PAH rats, and the co-localization of FUT8 with α-SMA was also observed. SiRNA was used to knockdown FUT8 in PASMCs (siFUT8). After effectively silencing FUT8 expression, phenotypic changes induced in PASMCs by PDGF-BB stimulation were alleviated. FUT8 activated the AKT pathway, while the admission of AKT activator SC79 could partially counteract the negative effect of siFUT8 on the proliferation, apoptotic resistance, and phenotypic switching of PASMCs, which may be involved in the core fucosylation of vascular endothelial growth factor receptor (VEGFR). Our research confirmed the critical role of FUT8 and its mediated core fucosylation in pulmonary vascular remodeling in PAH, providing a potential novel therapeutic target for PAH.
Collapse
Affiliation(s)
- Wen Zhang
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wenchao Lin
- Department of nephrology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaofang Zeng
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Mengqiu Zhang
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qin Chen
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yiyang Tang
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jing Sun
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Benhui Liang
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lihuang Zha
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders (Xiang Ya), Changsha, Hunan, China
| | - Zaixin Yu
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders (Xiang Ya), Changsha, Hunan, China
| |
Collapse
|
26
|
Chan BD, Wong WY, Lee MML, Yue PYK, Dai X, Tsim KWK, Hsiao WLW, Li M, Li XY, Tai WCS. Isolation and characterization of ZK002, a novel dual function snake venom protein from Deinagkistrodon acutus with anti-angiogenic and anti-inflammatory properties. Front Pharmacol 2023; 14:1227962. [PMID: 37841933 PMCID: PMC10570812 DOI: 10.3389/fphar.2023.1227962] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 09/19/2023] [Indexed: 10/17/2023] Open
Abstract
Introduction: Pathological angiogenesis, the abnormal or excessive generation of blood vessels, plays an important role in many diseases including cancer, diabetic retinopathy, psoriasis, and arthritis. Additionally, increasing evidence supports the close linkage between angiogenesis and inflammation. Snake venoms are a rich natural source of biologically active molecules and carry rich potential for the discovery of anti-angiogenic and anti-inflammatory modulators. Methods: Here, we isolated and purified a novel protein, ZK002, from the venom of the snake Deinagkistrodon acutus, and investigated its anti-angiogenic and anti-inflammatory activities and mechanisms. Results: ZK002 was identified as a 30 kDa heterodimeric protein of α and β chains, which exhibited anti-angiogenic activity in various in vitro assays. Mechanistically, ZK002 inhibited activation of VEGF signaling and related mediators including eNOS, p38, LIMK, and HSP27. ZK002 also upregulated the metalloproteinase inhibitor TIMP3 and inhibited components of the VEGF-induced signaling cascade, PPP3R2 and SH2D2A. The anti-angiogenic activity of ZK002 was confirmed in multiple in vivo models. ZK002 could also inhibit the in vitro expression of pro-inflammatory cytokines, as well as in vivo inflammation in the carrageenin-induced edema rat model. Conclusion: Our findings highlight the potential for further development of ZK002 as a dual function therapeutic against diseases with involvement of pathogenic angiogenesis and chronic inflammation.
Collapse
Affiliation(s)
- Brandon Dow Chan
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Wing-Yan Wong
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Magnolia Muk-Lan Lee
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Patrick Ying-Kit Yue
- Department of Biology, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
| | - Xiangrong Dai
- Lee’s Pharmaceutical (HK) Limited, Hong Kong Science Park, Shatin, Hong Kong SAR, China
| | - Karl Wah-Keung Tsim
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR, China
| | - Wen-Luan Wendy Hsiao
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Hong Kong SAR, China
| | - Mandy Li
- Zhaoke (Hong Kong) Ophthalmology Pharmaceutical Limited, Hong Kong Science Park, Shatin, Hong Kong SAR, China
| | - Xiao-Yi Li
- Zhaoke (Hong Kong) Ophthalmology Pharmaceutical Limited, Hong Kong Science Park, Shatin, Hong Kong SAR, China
| | - William Chi-Shing Tai
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), Shenzhen Research Institute of the Hong Kong Polytechnic University, Shenzhen, Hong Kong SAR, China
| |
Collapse
|
27
|
Lin PK, Davis GE. Extracellular Matrix Remodeling in Vascular Disease: Defining Its Regulators and Pathological Influence. Arterioscler Thromb Vasc Biol 2023; 43:1599-1616. [PMID: 37409533 PMCID: PMC10527588 DOI: 10.1161/atvbaha.123.318237] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 06/23/2023] [Indexed: 07/07/2023]
Abstract
Because of structural and cellular differences (ie, degrees of matrix abundance and cross-linking, mural cell density, and adventitia), large and medium-sized vessels, in comparison to capillaries, react in a unique manner to stimuli that induce vascular disease. A stereotypical vascular injury response is ECM (extracellular matrix) remodeling that occurs particularly in larger vessels in response to injurious stimuli, such as elevated angiotensin II, hyperlipidemia, hyperglycemia, genetic deficiencies, inflammatory cell infiltration, or exposure to proinflammatory mediators. Even with substantial and prolonged vascular damage, large- and medium-sized arteries, persist, but become modified by (1) changes in vascular wall cellularity; (2) modifications in the differentiation status of endothelial cells, vascular smooth muscle cells, or adventitial stem cells (each can become activated); (3) infiltration of the vascular wall by various leukocyte types; (4) increased exposure to critical growth factors and proinflammatory mediators; and (5) marked changes in the vascular ECM, that remodels from a homeostatic, prodifferentiation ECM environment to matrices that instead promote tissue reparative responses. This latter ECM presents previously hidden matricryptic sites that bind integrins to signal vascular cells and infiltrating leukocytes (in coordination with other mediators) to proliferate, invade, secrete ECM-degrading proteinases, and deposit injury-induced matrices (predisposing to vessel wall fibrosis). In contrast, in response to similar stimuli, capillaries can undergo regression responses (rarefaction). In summary, we have described the molecular events controlling ECM remodeling in major vascular diseases as well as the differential responses of arteries versus capillaries to key mediators inducing vascular injury.
Collapse
Affiliation(s)
- Prisca K. Lin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL 33612
| | - George E. Davis
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL 33612
| |
Collapse
|
28
|
Li S, Hu W, Gong S, Zhang P, Cheng J, Wang S, Wang Y, Shi W, Li Q, Wang F, Yuan Z. The Role of PRRC2B in Cerebral Vascular Remodeling Under Acute Hypoxia in Mice. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300892. [PMID: 37395402 PMCID: PMC10477837 DOI: 10.1002/advs.202300892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 05/13/2023] [Indexed: 07/04/2023]
Abstract
High altitude exposure leads to various cognitive impairments. The cerebral vasculature system plays an integral role in hypoxia-induced cognitive defects by reducing oxygen and nutrition supply to the brain. RNA N6-methyladenosine (m6A) is susceptible to modification and regulates gene expression in response to environmental changes, including hypoxia. However, the biological significance of m6A in endothelial cell performance under hypoxic conditions is unknown. Using m6A-seq, RNA immunoprcipitation-seq, and transcriptomic co-analysis, the molecular mechanism of vascular system remodeling under acute hypoxia is investigated. A novel m6A reader protein, proline-rich coiled-coil 2B (PRRC2B), exists in endothelial cells. PRRC2B knockdown promoted hypoxia-induced endothelial cell migration by regulating alternative splicing of the alpha 1 chain of collagen type XII in an m6A-dependent manner and the decay of matrix metallopeptidase domain 14 and ADAM metallopeptidase domain 19 mRNA in an m6A-independent manner. In addition, conditional knockout of PRRC2B in endothelial cells promotes hypoxia-induced vascular remodeling and cerebral blood flow redistribution, thus alleviating hypoxia-induced cognitive decline. Therefore, PRRC2B is integral in the hypoxia-induced vascular remodeling process as a novel RNA-binding protein. These findings provide a new potential therapeutic target for hypoxia-induced cognitive decline.
Collapse
Affiliation(s)
- Shuoshuo Li
- School of Life ScienceBeijing University of Chinese MedicineBeijing100029China
- The Brain Science CenterBeijing Institute of Basic Medical SciencesBeijing100850China
| | - Wenyu Hu
- The Brain Science CenterBeijing Institute of Basic Medical SciencesBeijing100850China
- School of MedicineUniversity of South ChinaHengyang421001China
| | - Shenghui Gong
- The Brain Science CenterBeijing Institute of Basic Medical SciencesBeijing100850China
| | - Ping Zhang
- The Brain Science CenterBeijing Institute of Basic Medical SciencesBeijing100850China
- School of MedicineUniversity of South ChinaHengyang421001China
| | - Jinbo Cheng
- The Brain Science CenterBeijing Institute of Basic Medical SciencesBeijing100850China
- Center on Translational NeuroscienceCollege of Life & Environmental ScienceMinzu University of ChinaBeijing100081China
| | - Shukun Wang
- The Brain Science CenterBeijing Institute of Basic Medical SciencesBeijing100850China
| | - Yingyi Wang
- The Brain Science CenterBeijing Institute of Basic Medical SciencesBeijing100850China
| | - Wenjun Shi
- The Brain Science CenterBeijing Institute of Basic Medical SciencesBeijing100850China
- Center on Translational NeuroscienceCollege of Life & Environmental ScienceMinzu University of ChinaBeijing100081China
| | - Qianqian Li
- The Brain Science CenterBeijing Institute of Basic Medical SciencesBeijing100850China
| | - Fengchao Wang
- National Institute of Biological SciencesBeijing102206China
| | - Zengqiang Yuan
- The Brain Science CenterBeijing Institute of Basic Medical SciencesBeijing100850China
| |
Collapse
|
29
|
Doherty EL, Aw WY, Warren EC, Hockenberry M, Whitworth CP, Krohn G, Howell S, Diekman BO, Legant WR, Nia HT, Hickey AJ, Polacheck WJ. Patient-derived extracellular matrix demonstrates role of COL3A1 in blood vessel mechanics. Acta Biomater 2023; 166:346-359. [PMID: 37187299 PMCID: PMC10330735 DOI: 10.1016/j.actbio.2023.05.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 05/17/2023]
Abstract
Vascular Ehlers-Danlos Syndrome (vEDS) is a rare autosomal dominant disease caused by mutations in the COL3A1 gene, which renders patients susceptible to aneurysm and arterial dissection and rupture. To determine the role of COL3A1 variants in the biochemical and biophysical properties of human arterial ECM, we developed a method for synthesizing ECM directly from vEDS donor fibroblasts. We found that the protein content of the ECM generated from vEDS donor fibroblasts differed significantly from ECM from healthy donors, including upregulation of collagen subtypes and other proteins related to ECM structural integrity. We further found that ECM generated from a donor with a glycine substitution mutation was characterized by increased glycosaminoglycan content and unique viscoelastic mechanical properties, including increased time constant for stress relaxation, resulting in a decrease in migratory speed of human aortic endothelial cells when seeded on the ECM. Collectively, these results demonstrate that vEDS patient-derived fibroblasts harboring COL3A1 mutations synthesize ECM that differs in composition, structure, and mechanical properties from healthy donors. These results further suggest that ECM mechanical properties could serve as a prognostic indicator for patients with vEDS, and the insights provided by the approach demonstrate the broader utility of cell-derived ECM in disease modeling. STATEMENT OF SIGNIFICANCE: The role of collagen III ECM mechanics remains unclear, despite reported roles in diseases including fibrosis and cancer. Here, we generate fibrous, collagen-rich ECM from primary donor cells from patients with vascular Ehlers-Danlos syndrome (vEDS), a disease caused by mutations in the gene that encodes collagen III. We observe that ECM grown from vEDS patients is characterized by unique mechanical signatures, including altered viscoelastic properties. By quantifying the structural, biochemical, and mechanical properties of patient-derived ECM, we identify potential drug targets for vEDS, while defining a role for collagen III in ECM mechanics more broadly. Furthermore, the structure/function relationships of collagen III in ECM assembly and mechanics will inform the design of substrates for tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Elizabeth L Doherty
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, NC, USA; UNC Catalyst for Rare Diseases, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Wen Yih Aw
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, NC, USA; UNC Catalyst for Rare Diseases, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Emily C Warren
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, NC, USA
| | - Max Hockenberry
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA; Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Chloe P Whitworth
- Department of Genetics, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Grace Krohn
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, NC, USA
| | - Stefanie Howell
- UNC Catalyst for Rare Diseases, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Brian O Diekman
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, NC, USA
| | - Wesley R Legant
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, NC, USA; Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Hadi Tavakoli Nia
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Anthony J Hickey
- UNC Catalyst for Rare Diseases, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - William J Polacheck
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, NC, USA; Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA; McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
30
|
Rusu CC, Kacso I, Moldovan D, Potra A, Tirinescu D, Ticala M, Rotar AM, Orasan R, Budurea C, Barar A, Anton F, Valea A, Bondor CI, Ticolea M. Triiodothyronine and Protein Malnutrition Could Influence Pulse Wave Velocity in Pre-Dialysis Chronic Kidney Disease Patients. Diagnostics (Basel) 2023; 13:2462. [PMID: 37510208 PMCID: PMC10377851 DOI: 10.3390/diagnostics13142462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/15/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023] Open
Abstract
Cardiovascular diseases (CVD) are the first cause of chronic kidney disease (CKD) mortality. For personalized improved medicine, detecting correctable markers of CVD can be considered a priority. The aim of this study was the evaluation of the impact of nutritional, hormonal and inflammatory markers on brachial-ankle Pulse Wave Velocity (PWV) in pre-dialysis CKD patients. A cross-sectional observational study was conducted on 68 pre-dialysis CKD patients (median age of 69 years, 41.2% with diabetes mellitus, 52.9% male). Laboratory data were collected, including levels of prolactin, triiodothyronine, TGF α, IL-6, and IL-1β. The high values of brachial-ankle PWV were associated with reduced muscle mass (p = 0.001, r = -0.44), low levels of total cholesterol (p = 0.04, r = -0.26), triglycerides (p = 0.03, r = -0.31), triiodothyronine (p = 0.04, r = -0.24), and prolactin (p = 0.02, r = -0.27). High PWV was associated with advanced age (p < 0.001, r = 0.19). In the multivariate analysis, reduced muscle mass (p = 0.018), low levels of triiodothyronine (p = 0.002), and triglycerides (p = 0.049) were significant predictors of PWV, but age (p < 0.001) remained an important factor. In conclusion, reduced triiodothyronine together with markers of malnutrition and age were associated with PWV in pre-dialysis CKD patients.
Collapse
Affiliation(s)
- Crina Claudia Rusu
- Department of Nephrology, University of Medicine and Pharmacy "Iuliu Hatieganu" Cluj, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania
- Department of Nephrology, County Emergency Clinical Hospital Cluj, 3-5 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Ina Kacso
- Department of Nephrology, University of Medicine and Pharmacy "Iuliu Hatieganu" Cluj, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania
- Department of Nephrology, County Emergency Clinical Hospital Cluj, 3-5 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Diana Moldovan
- Department of Nephrology, University of Medicine and Pharmacy "Iuliu Hatieganu" Cluj, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania
- Department of Nephrology, County Emergency Clinical Hospital Cluj, 3-5 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Alina Potra
- Department of Nephrology, University of Medicine and Pharmacy "Iuliu Hatieganu" Cluj, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania
- Department of Nephrology, County Emergency Clinical Hospital Cluj, 3-5 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Dacian Tirinescu
- Department of Nephrology, University of Medicine and Pharmacy "Iuliu Hatieganu" Cluj, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania
- Department of Nephrology, County Emergency Clinical Hospital Cluj, 3-5 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Maria Ticala
- Department of Nephrology, University of Medicine and Pharmacy "Iuliu Hatieganu" Cluj, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania
- Department of Nephrology, County Emergency Clinical Hospital Cluj, 3-5 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Ancuta M Rotar
- Department of Food Science, Faculty of Food Science and Technology, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, Calea Manastur 3-5, 400372 Cluj-Napoca, Romania
| | - Remus Orasan
- Nefromed Dialysis Center, 40 Ana Aslan Street, 400528 Cluj-Napoca, Romania
| | - Cristian Budurea
- Nefromed Dialysis Center, 40 Ana Aslan Street, 400528 Cluj-Napoca, Romania
| | - Andrada Barar
- Department of Nephrology, County Emergency Clinical Hospital Cluj, 3-5 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Florin Anton
- Department of Cardiology, University of Medicine and Pharmacy "Iuliu Hatieganu" Cluj, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania
| | - Ana Valea
- Department of Endocrinology, University of Medicine and Pharmacy "Iuliu Hatieganu" Cluj, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania
| | - Cosmina Ioana Bondor
- Department of Medical Informatics and Biostatistics, University of Medicine and Pharmacy "Iuliu Hatieganu" Cluj, 6 Pasteur Street, 400349 Cluj-Napoca, Romania
| | - Madalina Ticolea
- Department of Nephrology, County Emergency Clinical Hospital Cluj, 3-5 Clinicilor Street, 400006 Cluj-Napoca, Romania
| |
Collapse
|
31
|
Chang C, Guo W, Yu X, Guo C, Zhou N, Guo X, Huang RL, Li Q, Zhu Y. Engineered M13 phage as a novel therapeutic bionanomaterial for clinical applications: From tissue regeneration to cancer therapy. Mater Today Bio 2023; 20:100612. [PMID: 37063776 PMCID: PMC10102448 DOI: 10.1016/j.mtbio.2023.100612] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/12/2023] [Accepted: 03/15/2023] [Indexed: 04/03/2023] Open
Abstract
Bacteriophages (phages) are nanostructured viruses with highly selective antibacterial properties that have gained attention beyond eliminating bacteria. Specifically, M13 phages are filamentous phages that have recently been studied in various aspects of nanomedicine due to their biological advantages and more compliant engineering capabilities over other phages. Having nanofiber-like morphology, M13 phages can reach varied target sites and self-assemble into multidimensional scaffolds in a relatively safe and stable way. In addition, genetic modification of the coat proteins enables specific display of peptides and antibodies on the phages, allowing for precise and individualized medicine. M13 phages have also been subjected to novel engineering approaches, including phage-based bionanomaterial engineering and phage-directed nanomaterial combinations that enhance the bionanomaterial properties of M13 phages. In view of these features, researchers have been able to utilize M13 phages for therapeutic applications such as drug delivery, biodetection, tissue regeneration, and targeted cancer therapy. In particular, M13 phages have been utilized as a novel bionanomaterial for precisely mimicking natural tissue environment in order to overcome the shortage in tissue and organ donors. Hence, in this review, we address the recent studies and advances of using M13 phages in the field of nanomedicine as therapeutic agents based upon their characteristics as novel bionanomaterial with biomolecules displayed. This paper also emphasizes the novel engineering approach that enhances M13 phage's bionanomaterial capabilities. Current limitations and future approaches are also discussed to provide insight in further progress for M13 phage-based clinical applications.
Collapse
Affiliation(s)
- Cheng Chang
- School of Global Health, Chinese Centre for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, One Health Center, Shanghai Jiao Tong University-The University of Edinburgh, Shanghai, 200025, China
| | - Wennan Guo
- School of Global Health, Chinese Centre for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, One Health Center, Shanghai Jiao Tong University-The University of Edinburgh, Shanghai, 200025, China
| | - Xinbo Yu
- Second Dental Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201999, China
| | - Chaoyi Guo
- School of Global Health, Chinese Centre for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, One Health Center, Shanghai Jiao Tong University-The University of Edinburgh, Shanghai, 200025, China
| | - Nan Zhou
- School of Global Health, Chinese Centre for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, One Health Center, Shanghai Jiao Tong University-The University of Edinburgh, Shanghai, 200025, China
| | - Xiaokui Guo
- School of Global Health, Chinese Centre for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, One Health Center, Shanghai Jiao Tong University-The University of Edinburgh, Shanghai, 200025, China
| | - Ru-Lin Huang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Corresponding author.
| | - Qingtian Li
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Corresponding author.
| | - Yongzhang Zhu
- School of Global Health, Chinese Centre for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, One Health Center, Shanghai Jiao Tong University-The University of Edinburgh, Shanghai, 200025, China
- Corresponding author.
| |
Collapse
|
32
|
Gao J, Li L, Zhou D, Sun X, Cui L, Yang D, Wang X, Du P, Yuan W. Effects of norepinephrine‑induced activation of rat vascular adventitial fibroblasts on proliferation and migration of BMSCs involved in vascular remodeling. Exp Ther Med 2023; 25:290. [PMID: 37206559 PMCID: PMC10189611 DOI: 10.3892/etm.2023.11989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 04/11/2023] [Indexed: 05/21/2023] Open
Abstract
Vascular remodeling caused by vascular injury such as hypertension and atherosclerosis is a complex process involving a variety of cells and factors, and the mechanism is unclear. A vascular injury model was simulated by adding norepinephrine (NE) to culture medium of vascular adventitial fibroblasts (AFs). NE induced activation and proliferation of AFs. To investigate the association between the AFs activation and bone marrow mesenchymal stem cells (BMSCs) differentiation in vascular remodeling. BMSCs were cultured with supernatant of the AFs culture medium. BMSC differentiation and migration were observed by immunostaining and Transwell assay, respectively, while cell proliferation was measured using the Cell Counting Kit-8. Expression levels of smooth muscle actin (α-SMA), TGF-β1 and SMAD3 were measured using western blot assay. The results indicated that compared with those in the control group, in which BMSCs were cultured in normal medium, expression levels of α-SMA, TGF-β1 and SMAD3 in BMSCs cultured in medium supplemented with supernatant of AFs, increased significantly (all P<0.05). Activated AFs induced the differentiation of BMSCs into vascular smooth muscle-like cells and promoted proliferation and migration. AFs activated by NE may induce BMSCs to participate in vascular remodeling. These findings may help design and develop new approaches and therapeutic strategies for vascular injury to prevent pathological remodeling.
Collapse
Affiliation(s)
- Jun Gao
- Medical Laboratory, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong 264100, P.R. China
| | - Li Li
- Pediatric Department, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong 264100, P.R. China
| | - Dongli Zhou
- Nurse's Office, Health School of Laiyang, Laiyang, Yantai, Shandong 265200, P.R. China
| | - Xuhong Sun
- Institute of Pathology and Pathophysiology, Basic Medical School, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Lilu Cui
- Institute of Pathology and Pathophysiology, Basic Medical School, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Donglin Yang
- Institute of Pathology and Pathophysiology, Basic Medical School, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Xiaohui Wang
- Institute of Pathology and Pathophysiology, Basic Medical School, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Pengchao Du
- Institute of Pathology and Pathophysiology, Basic Medical School, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
- Correspondence to: Professor Wendan Yuan or Professor Pengchao Du, Institute of Pathology and Pathophysiology, Basic Medical School, Binzhou Medical University, 346 Guanhai Road, Yantai, Shandong 264003, P.R. China E-mail: 981713509 @qq.com
| | - Wendan Yuan
- Institute of Pathology and Pathophysiology, Basic Medical School, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
- Correspondence to: Professor Wendan Yuan or Professor Pengchao Du, Institute of Pathology and Pathophysiology, Basic Medical School, Binzhou Medical University, 346 Guanhai Road, Yantai, Shandong 264003, P.R. China E-mail: 981713509 @qq.com
| |
Collapse
|
33
|
Ding R, Ren X, Sun Q, Sun Z, Duan J. An integral perspective of canonical cigarette and e-cigarette-related cardiovascular toxicity based on the adverse outcome pathway framework. J Adv Res 2023; 48:227-257. [PMID: 35998874 PMCID: PMC10248804 DOI: 10.1016/j.jare.2022.08.012] [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: 06/11/2022] [Revised: 07/29/2022] [Accepted: 08/15/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Nowadays, cigarette smoking remains the leading cause of chronic disease and premature death, especially cardiovascular disease. As an emerging tobacco product, e-cigarettes have been advocated as alternatives to canonical cigarettes, and thus may be an aid to promote smoking cessation. However, recent studies indicated that e-cigarettes should not be completely harmless to the cardiovascular system. AIM OF REVIEW This review aimed to build up an integral perspective of cigarettes and e-cigarettes-related cardiovascular toxicity. KEY SCIENTIFIC CONCEPTS OF REVIEW This review adopted the adverse outcome pathway (AOP) framework as a pivotal tool and aimed to elucidate the association between the molecular initiating events (MIEs) induced by cigarette and e-cigarette exposure to the cardiovascular adverse outcome. Since the excessive generation of reactive oxygen species (ROS) has been widely approved to play a critical role in cigarette smoke-related CVD and may also be involved in e-cigarette-induced toxic effects, the ROS overproduction and subsequent oxidative stress are regarded as essential parts of this framework. As far as we know, this should be the first AOP framework focusing on cigarette and e-cigarette-related cardiovascular toxicity, and we hope our work to be a guide in exploring the biomarkers and novel therapies for cardiovascular injury.
Collapse
Affiliation(s)
- Ruiyang Ding
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Xiaoke Ren
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Qinglin Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China.
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China.
| |
Collapse
|
34
|
Fu Y, Huang J, He S, Yan X, Wang X, Lian H, Zeng Y, Li D, Guo R. Betulinaldehyde inhibits vascular remodeling by regulating the microenvironment through the PLCγ1/Ca 2+/MMP9 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 116:154891. [PMID: 37229891 DOI: 10.1016/j.phymed.2023.154891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 05/02/2023] [Accepted: 05/16/2023] [Indexed: 05/27/2023]
Abstract
BACKGROUND Vascular remodeling plays a crucial role in the pathogenesis of several cardiovascular diseases (CVDs). Unfortunately, current drug therapies offer limited relief for vascular remodeling. Therefore, the development of innovative therapeutic strategies or drugs that target vascular remodeling is imperative. Betulinaldehyde (BA) is a triterpenoid with diverse biological activities, but its effects on vascular remodeling remain unclear. OBJECTIVE This study aimed to investigate the role of BA in vascular remodeling and its mechanism of action, providing valuable information for future applications of BA in the treatment of CVDs. METHODS Network pharmacology was used to predict the key targets of BA in vascular remodeling. The effect of BA on vascular remodeling was assessed in a rat model of balloon injury using hematoxylin and eosin staining, Masson staining, immunohistochemistry staining, and Western blotting. A phenotypic transformation model of vascular smooth muscle cells (VSMCs) was induced by platelet-derived growth factor-BB, and the functional impacts of BA on VSMCs were assessed via CCK-8, EdU, Wound healing, Transwell, and Western blotting. Finally, after manipulation of phospholipase C gamma1 (PLCγ1) expression, Western blotting and Ca2+ levels determination were performed to investigate the potential mechanism of action of BA. RESULTS The most key target of BA in vascular remodeling, matrix metalloproteinase 9 (MMP9), was identified through network pharmacology screening. Vascular remodeling was alleviated by BA in vivo and its effects were associated with decreased MMP9 expression. In vitro studies indicated that BA inhibited VSMC proliferation, migration, phenotypic transformation, and downregulated MMP9 expression. Additionally, BA decreased PLCγ1 expression and Ca2+ levels in VSMCs. However, after pretreatment with a phospholipase C agonist, BA's effects on down-regulating the expression of PLCγ1 and Ca2+ levels were inhibited, while the expression of MMP9 increased compared to that in the BA treatment group. CONCLUSION This study demonstrated the critical role of BA in vascular remodeling. These findings revealed a novel mechanism whereby BA mediates its protective effects through MMP9 regulation by inhibiting the PLCγ1/Ca2+/MMP9 signaling pathway. Overall, BA may potentially be developed into a novel medication for CVDs and may serve as a promising therapeutic strategy for improving recovery from CVDs by targeting MMP9.
Collapse
Affiliation(s)
- Yangxia Fu
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Jun Huang
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Shuangyan He
- Department of Laboratory Animals, Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Xin Yan
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Xia Wang
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Huilin Lian
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Youjie Zeng
- Department of Anesthesiology, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Dai Li
- Phase I Clinical Trial Center, The Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Ren Guo
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China.
| |
Collapse
|
35
|
Molecular and Cellular Regulations in the Development of the Choroidal Circulation System. Int J Mol Sci 2023; 24:ijms24065371. [PMID: 36982446 PMCID: PMC10048934 DOI: 10.3390/ijms24065371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/09/2023] [Accepted: 03/09/2023] [Indexed: 03/18/2023] Open
Abstract
Disorders in the development and regulation of blood vessels are involved in various ocular disorders, such as persistent hyperplastic primary vitreous, familial exudative vitreoretinopathy, and choroidal dystrophy. Thus, the appropriate regulation of vascular development is essential for healthy ocular functions. However, regulation of the developing choroidal circulation system has not been well studied compared with vascular regulation in the vitreous and the retina. The choroid is a vascular-rich and uniquely structured tissue supplying oxygen and nutrients to the retina, and hypoplasia and the degeneration of the choroid are involved in many ocular disorders. Therefore, understanding the developing choroidal circulation system expands our knowledge of ocular development and supports our understanding of ocular disorders. In this review, we examine studies on regulating the developing choroidal circulation system at the cellular and molecular levels and discuss the relevance to human diseases.
Collapse
|
36
|
Gong Z, Huang J, Wang D, Yang S, Ma Z, Fu Y, Ma Q, Kong W. ADAMTS-7 deficiency attenuates thoracic aortic aneurysm and dissection in mice. J Mol Med (Berl) 2023; 101:237-248. [PMID: 36662289 DOI: 10.1007/s00109-023-02284-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/18/2022] [Accepted: 01/10/2023] [Indexed: 01/21/2023]
Abstract
Thoracic aortic aneurysm and dissection (TAAD) is a life-threatening cardiovascular disease with severe extracellular matrix (ECM) remodeling that lacks efficient early stage diagnosis and nonsurgical therapy. A disintegrin and metalloproteinase with thrombospondin motif 7 (ADAMTS-7) is recognized as a novel locus for human coronary artery atherosclerosis. Previous work by us and others showed that ADAMTS-7 promoted atherosclerosis, postinjury neointima formation, and vascular calcification. However, whether ADAMTS-7 is involved in TAAD pathogenesis is unknown. We aimed to explore the alterations in ADAMTS-7 expression in human and mouse TAAD, and investigate the role of ADAMTS-7 in TAAD formation. A case-control study of TAAD patients (N = 86) and healthy participants (N = 88) was performed. The plasma ADAMTS-7 levels were markedly increased in TAAD patients within 24 h and peaked in 7 days. A TAAD mouse model was induced with 0.5% β-aminopropionitrile (BAPN) in drinking water. ELISA analysis of mouse plasma, Western blotting, and immunohistochemical staining of aorta showed an increase in ADAMTS-7 in the early stage of TAAD. Moreover, ADAMTS-7-deficient mice exhibited significantly attenuated TAAD formation and TAAD rupture-related mortality in both male and female mice, which was accompanied by reduced artery dilation and inhibited elastin degradation. ADAMTS-7 deficiency caused repressed inflammatory response and complement system activation during TAAD formation. An increase in plasma ADAMTS-7 is a novel biomarker for human TAAD. ADAMTS-7 deficiency attenuates BAPN-induced murine TAAD. ADAMTS-7 is a potential novel target for TAAD diagnosis and therapy. KEY MESSAGES: A case-control study revealed increased plasma ADAMTS-7 is a risk factor for TAAD. ADAMTS-7 was elevated in plasma and aorta at early stage of mouse TAAD. ADAMTS-7 knockout attenuated mouse TAAD formation and mortality in both sexes.
Collapse
Affiliation(s)
- Ze Gong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, 100191, People's Republic of China
| | - Jiaqi Huang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, 100191, People's Republic of China
| | - Daidai Wang
- Department of Emergency Medicine, Peking University Third Hospital, Beijing, 100191, People's Republic of China
| | - Shiyu Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, 100191, People's Republic of China
| | - Zihan Ma
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, 100191, People's Republic of China
| | - Yi Fu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, 100191, People's Republic of China
| | - Qingbian Ma
- Department of Emergency Medicine, Peking University Third Hospital, Beijing, 100191, People's Republic of China.
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, 100191, People's Republic of China.
| |
Collapse
|
37
|
Ma Z, Mao C, Chen X, Yang S, Qiu Z, Yu B, Jia Y, Wu C, Wang Y, Wang Y, Gu R, Yu F, Yin Y, Wang X, Xu Q, Liu C, Liao Y, Zheng J, Fu Y, Kong W. Peptide Vaccine Against ADAMTS-7 Ameliorates Atherosclerosis and Postinjury Neointima Hyperplasia. Circulation 2023; 147:728-742. [PMID: 36562301 DOI: 10.1161/circulationaha.122.061516] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND The metalloprotease ADAMTS-7 (a disintegrin and metalloproteinase with thrombospondin type 1 motif 7) is a novel locus associated with human coronary atherosclerosis. ADAMTS-7 deletion protects against atherosclerosis and vascular restenosis in rodents. METHODS We designed 3 potential vaccines consisting of distinct B cell epitopic peptides derived from ADAMTS-7 and conjugated with the carrier protein KLH (keyhole limpet hemocyanin) as well as aluminum hydroxide as an adjuvant. Arterial ligation or wire injury was used to induce neointima in mice, whereas ApoE-/- and LDLR-/- (LDLR [low-density lipoprotein receptor]) mice fed a high-fat diet were applied to assess atherosclerosis. In addition, coronary stent implantation was performed on vaccine-immunized Bama miniature pigs, followed by optical coherence tomography to evaluate coronary intimal hyperplasia. RESULTS A vaccine, ATS7vac, was screened out from 3 candidates to effectively inhibit intimal thickening in murine carotid artery ligation models after vaccination. As well, immunization with ATS7vac alleviated neointima formation in murine wire injury models and mitigated atherosclerotic lesions in both hyperlipidemic ApoE-/- and LDLR-/- mice without lowering lipid levels. Preclinically, ATS7vac markedly impeded intimal hyperplasia in swine stented coronary arteries, but without significant immune-related organ injuries. Mechanistically, ATS7vac vaccination produced specific antibodies against ADAMTS-7, which markedly repressed ADAMTS-7-mediated COMP (cartilage oligomeric matrix protein) and TSP-1 (thrombospondin-1) degradation and subsequently inhibited vascular smooth muscle cell migration but promoted re-endothelialization. CONCLUSIONS ATS7vac is a novel atherosclerosis vaccine that also alleviates in-stent restenosis. The application of ATS7vac would be a complementary therapeutic avenue to the current lipid-lowering strategy for atherosclerotic disease.
Collapse
Affiliation(s)
- Zihan Ma
- Department of Physiology and Pathophysiology (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Ministry of Education, Beijing, China
| | - Chenfeng Mao
- Department of Physiology and Pathophysiology (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Ministry of Education, Beijing, China.,Beijing Institute of Biotechnology, Beijing, China (C.M.)
| | - Xiao Chen
- Department of Cardiology and Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (X.C., Z.Q., Yiyi Wang, Y.L.).,Key Laboratory of Molecular Biological Targeted Therapies, Ministry of Education, Wuhan, China (X.C., Z.Q., Yiyi Wang, Y.L.)
| | - Shiyu Yang
- Department of Physiology and Pathophysiology (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Ministry of Education, Beijing, China
| | - Zhihua Qiu
- Department of Cardiology and Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (X.C., Z.Q., Yiyi Wang, Y.L.).,Key Laboratory of Molecular Biological Targeted Therapies, Ministry of Education, Wuhan, China (X.C., Z.Q., Yiyi Wang, Y.L.)
| | - Baoqi Yu
- Key Laboratory of Remodeling-Related Cardiovascular Diseases (B.Y.), Ministry of Education, Beijing, China.,Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China (B.Y.)
| | - Yiting Jia
- Department of Physiology and Pathophysiology (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Ministry of Education, Beijing, China
| | - Chao Wu
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China (C.W., J.Z.)
| | - Yiyi Wang
- Department of Cardiology and Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (X.C., Z.Q., Yiyi Wang, Y.L.).,Key Laboratory of Molecular Biological Targeted Therapies, Ministry of Education, Wuhan, China (X.C., Z.Q., Yiyi Wang, Y.L.)
| | - Yuhui Wang
- School of Basic Medical Sciences, Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education (Yuhui Wang), Peking University, Beijing, China
| | - Rui Gu
- Department of Physiology and Pathophysiology (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Ministry of Education, Beijing, China
| | - Fang Yu
- Department of Physiology and Pathophysiology (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Ministry of Education, Beijing, China
| | - Yanhui Yin
- Department of Immunology (Y.Y.), Peking University, Beijing, China.,Key Laboratory of Medical Immunology of Ministry of Health (Y.Y.), Peking University, Beijing, China
| | - Xian Wang
- Department of Physiology and Pathophysiology (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Ministry of Education, Beijing, China
| | - Qingbo Xu
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre, United Kingdom (Q.X.).,Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China (Q.X.)
| | - Chuanju Liu
- Department of Orthopedic Surgery, Department of Cell Biology, New York University School of Medicine, New York, NY (C.L.)
| | - Yuhua Liao
- Department of Cardiology and Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (X.C., Z.Q., Yiyi Wang, Y.L.).,Key Laboratory of Molecular Biological Targeted Therapies, Ministry of Education, Wuhan, China (X.C., Z.Q., Yiyi Wang, Y.L.)
| | - Jingang Zheng
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China (C.W., J.Z.)
| | - Yi Fu
- Department of Physiology and Pathophysiology (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Ministry of Education, Beijing, China
| | - Wei Kong
- Department of Physiology and Pathophysiology (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Ministry of Education, Beijing, China
| |
Collapse
|
38
|
Tan JL, Yi J, Cao XY, Wang FY, Xie SL, Zhou LL, Qin L, Dai AG. Celastrol: The new dawn in the treatment of vascular remodeling diseases. Biomed Pharmacother 2023; 158:114177. [PMID: 36809293 DOI: 10.1016/j.biopha.2022.114177] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/16/2022] [Accepted: 12/28/2022] [Indexed: 01/05/2023] Open
Abstract
Evidence is mounting that abnormal vascular remodeling leads to many cardiovascular diseases (CVDs). This suggests that vascular remodeling can be a crucial target for the prevention and treatment of CVDs. Recently, celastrol, an active ingredient of the broadly used Chinese herb Tripterygium wilfordii Hook F, has attracted extensive interest for its proven potential to improve vascular remodeling. Substantial evidence has shown that celastrol improves vascular remodeling by ameliorating inflammation, hyperproliferation, and migration of vascular smooth muscle cells, vascular calcification, endothelial dysfunction, extracellular matrix remodeling, and angiogenesis. Moreover, numerous reports have proven the positive effects of celastrol and its therapeutic promise in treating vascular remodeling diseases such as hypertension, atherosclerosis, and pulmonary artery hypertension. The present review summarizes and discusses the molecular mechanism of celastrol regulating vascular remodeling and provides preclinical proof for future clinical applications of celastrol.
Collapse
Affiliation(s)
- Jun-Lan Tan
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China
| | - Jian Yi
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410021, Hunan, China
| | - Xian-Ya Cao
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China
| | - Fei-Ying Wang
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China
| | - Si-Lin Xie
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China
| | - Ling-Ling Zhou
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China
| | - Li Qin
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China; Laboratory of Stem Cell Regulation with Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China.
| | - Ai-Guo Dai
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China; Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410021, Hunan, China.
| |
Collapse
|
39
|
Déglise S, Bechelli C, Allagnat F. Vascular smooth muscle cells in intimal hyperplasia, an update. Front Physiol 2023; 13:1081881. [PMID: 36685215 PMCID: PMC9845604 DOI: 10.3389/fphys.2022.1081881] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/12/2022] [Indexed: 01/05/2023] Open
Abstract
Arterial occlusive disease is the leading cause of death in Western countries. Core contemporary therapies for this disease include angioplasties, stents, endarterectomies and bypass surgery. However, these treatments suffer from high failure rates due to re-occlusive vascular wall adaptations and restenosis. Restenosis following vascular surgery is largely due to intimal hyperplasia. Intimal hyperplasia develops in response to vessel injury, leading to inflammation, vascular smooth muscle cells dedifferentiation, migration, proliferation and secretion of extra-cellular matrix into the vessel's innermost layer or intima. In this review, we describe the current state of knowledge on the origin and mechanisms underlying the dysregulated proliferation of vascular smooth muscle cells in intimal hyperplasia, and we present the new avenues of research targeting VSMC phenotype and proliferation.
Collapse
Affiliation(s)
| | | | - Florent Allagnat
- Department of Vascular Surgery, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
40
|
Dong R, Li J, Jiang G, Han N, Zhang Y, Shi X. Novel immune cell infiltration-related biomarkers in atherosclerosis diagnosis. PeerJ 2023; 11:e15341. [PMID: 37151293 PMCID: PMC10158768 DOI: 10.7717/peerj.15341] [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: 02/09/2023] [Accepted: 04/12/2023] [Indexed: 05/09/2023] Open
Abstract
Background Immune cell infiltration (ICI) has a close relationship with the progression of atherosclerosis (AS). Therefore, the current study was aimed to explore the role of genes related to ICI and to investigate potential mechanisms in AS. Methods Single-sample gene set enrichment analysis (ssGSEA) was applied to explore immune infiltration in AS and controls. Genes related to immune infitration were mined by weighted gene co-expression network analysis (WGCNA). The function of those genes were analyzed by enrichment analyses of the Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO). The interactions among those genes were visualized in the protein-protein interaction (PPI) network, followed by identification of hub genes through Cytoscape software. A receiver operating characteristic (ROC) plot was generated to assess the performance of hub genes in AS diagnosis. The expressions of hub genes were measured by reverse transcription quantitative real-time PCR (RT-qPCR) in human leukemia monocyticcell line (THP-1) derived foam cells and macrophages, which mimic AS and control, respectively. Results We observed that the proportions of 27 immune cells were significantly elevated in AS. Subsequent integrative analyses of differential expression and WGCNA identified 99 immune cell-related differentially expressed genes (DEGs) between AS and control. Those DEGs were associated with tryptophan metabolism and extracellular matrix (ECM)-related functions. Moreover, by constructing the PPI network, we found 11 hub immune cell-related genes in AS. The expression pattern and receiver ROC analyses in two independent datasets showed that calsequestrin 2 (CASQ2), nexilin F-Actin binding protein (NEXN), matrix metallopeptidase 12 (MMP12), C-X-C motif chemokine ligand 10 (CXCL10), phospholamban (PLN), heme oxygenase 1 (HMOX1), ryanodine receptor 2 (RYR2), chitinase 3 like 1 (CHI3L1), matrix metallopeptidase 9 (MMP9), actin alpha cardiac muscle 1 (ACTC1) had good performance in distinguishing AS from control samples. Furthermore, those biomarkers were shown to be correlated with angiogenesis and immune checkpoints. In addition, we found 239 miRNAs and 47 transcription factor s (TFs), which may target those biomarkers and regulate their expressions. Finally, we found that RT-qPCR results were consistent with sequencing results.
Collapse
Affiliation(s)
- Ruoyu Dong
- Department of Vascular Surgery, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Jikuan Li
- Department of Vascular Surgery, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Guangwei Jiang
- Department of Vascular Surgery, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Ning Han
- Department of Neurointervention, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Yaochao Zhang
- Department of Cardiothoracic Surgery, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Xiaoming Shi
- Department of Vascular Surgery, Hebei General Hospital, Shijiazhuang, Hebei, China
| |
Collapse
|
41
|
Bi J, Duan Y, Wang M, He C, Li X, Zhang X, Tao Y, Du Y, Liu H. Deletion of large-conductance calcium-activated potassium channels promotes vascular remodelling through the CTRP7-mediated PI3K/Akt signaling pathway. Acta Biochim Biophys Sin (Shanghai) 2022; 54:1-11. [PMID: 36514218 PMCID: PMC10157624 DOI: 10.3724/abbs.2022179] [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: 11/25/2022] Open
Abstract
The large-conductance calcium-activated potassium (BK) channel is a critical regulator and potential therapeutic target of vascular tone and architecture, and abnormal expression or dysfunction of this channel is linked to many vascular diseases. Vascular remodelling is the early pathological basis of severe vascular diseases. Delaying the progression of vascular remodelling can reduce cardiovascular events, but the pathogenesis remains unclear. To clarify the role of BK channels in vascular remodelling, we use rats with BK channel α subunit knockout (BK α ‒/‒). The results show that BK α ‒/‒ rats have smaller inner and outer diameters, thickened aortic walls, increased fibrosis, and disordered elastic fibers of the aortas compared with WT rats. When the expression and function of BK α are inhibited in human umbilical arterial smooth muscle cells (HUASMCs), the expressions of matrix metalloproteinase 2 (MMP2), MMP9, and interleukin-6 are enhanced, while the expressions of smooth muscle cell contractile phenotype proteins are reduced. RNA sequencing, bioinformatics analysis and qPCR verification show that C1q/tumor necrosis factor-related protein 7 ( CTRP7) is the downstream target gene. Furthermore, except for that of MMPs, a similar pattern of IL-6, smooth muscle cell contractile phenotype proteins expression trend is observed after CTRP7 knockdown. Moreover, knockdown of both BK α and CTRP7 in HUASMCs activates PI3K/Akt signaling. Additionally, CTRP7 is expressed in vascular smooth muscle cells (VSMCs), and BK α deficiency activates the PI3K/Akt pathway by reducing CTRP7 level. Therefore, we first show that BK channel deficiency leads to vascular remodelling. The BK channel and CTRP7 may serve as potential targets for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Jing Bi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China.,Beijing Key Laboratory of Metabolic Disturbance Related Cardiovascular Disease, Beijing 100069, China
| | - Yanru Duan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China.,Beijing Key Laboratory of Metabolic Disturbance Related Cardiovascular Disease, Beijing 100069, China
| | - Meili Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China.,Beijing Key Laboratory of Metabolic Disturbance Related Cardiovascular Disease, Beijing 100069, China
| | - Chunyu He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China.,Beijing Key Laboratory of Metabolic Disturbance Related Cardiovascular Disease, Beijing 100069, China
| | - Xiaoyue Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China.,Beijing Key Laboratory of Metabolic Disturbance Related Cardiovascular Disease, Beijing 100069, China
| | - Xi Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China.,Beijing Key Laboratory of Metabolic Disturbance Related Cardiovascular Disease, Beijing 100069, China
| | - Yan Tao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China.,Beijing Key Laboratory of Metabolic Disturbance Related Cardiovascular Disease, Beijing 100069, China
| | - Yunhui Du
- Beijing Key Laboratory of Upper Airway Dysfunction-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Huirong Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China.,Beijing Key Laboratory of Metabolic Disturbance Related Cardiovascular Disease, Beijing 100069, China
| |
Collapse
|
42
|
Singh D, Rai V, Agrawal DK. Non-Coding RNAs in Regulating Plaque Progression and Remodeling of Extracellular Matrix in Atherosclerosis. Int J Mol Sci 2022; 23:13731. [PMID: 36430208 PMCID: PMC9692922 DOI: 10.3390/ijms232213731] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/31/2022] [Accepted: 11/05/2022] [Indexed: 11/09/2022] Open
Abstract
Non-coding RNAs (ncRNAs) regulate cell proliferation, migration, differentiation, inflammation, metabolism of clinically important biomolecules, and other cellular processes. They do not encode proteins but are involved in the regulatory network of various proteins that are directly related to the pathogenesis of diseases. Little is known about the ncRNA-associated mechanisms of atherosclerosis and related cardiovascular disorders. Remodeling of the extracellular matrix (ECM) is critical in the pathogenesis of atherosclerosis and related disorders; however, its regulatory proteins are the potential subjects to explore with special emphasis on epigenetic regulatory components. The activity of regulatory proteins involved in ECM remodeling is regulated by various ncRNA molecules, as evident from recent research. Thus, it is important to critically evaluate the existing literature to enhance the understanding of nc-RNAs-regulated molecular mechanisms regulating ECM components, remodeling, and progression of atherosclerosis. This is crucial since deregulated ECM remodeling contributes to atherosclerosis. Thus, an in-depth understanding of ncRNA-associated ECM remodeling may identify novel targets for the treatment of atherosclerosis and other cardiovascular diseases.
Collapse
Affiliation(s)
| | | | - Devendra K. Agrawal
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| |
Collapse
|
43
|
Liu SF, Nambiar Veetil N, Li Q, Kucherenko MM, Knosalla C, Kuebler WM. Pulmonary hypertension: Linking inflammation and pulmonary arterial stiffening. Front Immunol 2022; 13:959209. [PMID: 36275740 PMCID: PMC9579293 DOI: 10.3389/fimmu.2022.959209] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 09/14/2022] [Indexed: 11/13/2022] Open
Abstract
Pulmonary hypertension (PH) is a progressive disease that arises from multiple etiologies and ultimately leads to right heart failure as the predominant cause of morbidity and mortality. In patients, distinct inflammatory responses are a prominent feature in different types of PH, and various immunomodulatory interventions have been shown to modulate disease development and progression in animal models. Specifically, PH-associated inflammation comprises infiltration of both innate and adaptive immune cells into the vascular wall of the pulmonary vasculature—specifically in pulmonary vascular lesions—as well as increased levels of cytokines and chemokines in circulating blood and in the perivascular tissue of pulmonary arteries (PAs). Previous studies suggest that altered hemodynamic forces cause lung endothelial dysfunction and, in turn, adherence of immune cells and release of inflammatory mediators, while the resulting perivascular inflammation, in turn, promotes vascular remodeling and the progression of PH. As such, a vicious cycle of endothelial activation, inflammation, and vascular remodeling may develop and drive the disease process. PA stiffening constitutes an emerging research area in PH, with relevance in PH diagnostics, prognostics, and as a therapeutic target. With respect to its prognostic value, PA stiffness rivals the well-established measurement of pulmonary vascular resistance as a predictor of disease outcome. Vascular remodeling of the arterial extracellular matrix (ECM) as well as vascular calcification, smooth muscle cell stiffening, vascular wall thickening, and tissue fibrosis contribute to PA stiffening. While associations between inflammation and vascular stiffening are well-established in systemic vascular diseases such as atherosclerosis or the vascular manifestations of systemic sclerosis, a similar connection between inflammatory processes and PA stiffening has so far not been addressed in the context of PH. In this review, we discuss potential links between inflammation and PA stiffening with a specific focus on vascular calcification and ECM remodeling in PH.
Collapse
Affiliation(s)
- Shao-Fei Liu
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Netra Nambiar Veetil
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, German Heart Center, Berlin, Germany
| | - Qiuhua Li
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Mariya M. Kucherenko
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, German Heart Center, Berlin, Germany
- *Correspondence: Mariya M. Kucherenko,
| | - Christoph Knosalla
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, German Heart Center, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Wolfgang M. Kuebler
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
- German Center for Lung Research (DZL), Gießen, Germany
- The Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON, Canada
- Department of Surgery and Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
44
|
Xu H, He Y, Hong T, Bi C, Li J, Xia M. Piezo1 in vascular remodeling of atherosclerosis and pulmonary arterial hypertension: A potential therapeutic target. Front Cardiovasc Med 2022; 9:1021540. [PMID: 36247424 PMCID: PMC9557227 DOI: 10.3389/fcvm.2022.1021540] [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: 08/17/2022] [Accepted: 09/12/2022] [Indexed: 11/13/2022] Open
Abstract
Vascular remodeling (VR) is a structural and functional change of blood vessels to adapt to the changes of internal and external environment. It is one of the common pathological features of many vascular proliferative diseases. The process of VR is mainly manifested in the changes of vascular wall structure and function, including intimal hyperplasia, thickening or thinning of media, fibrosis of adventitia, etc. These changes are also the pathological basis of aging and various cardiovascular diseases. Mechanical force is the basis of cardiovascular biomechanics, and the newly discovered mechanical sensitive ion channel Piezo1 is widely distributed in the whole cardiovascular system. Studies have confirmed that Piezo1, a mechanically sensitive ion channel, plays an important role in cardiovascular remodeling diseases. This article reviews the molecular mechanism of Piezo1 in atherosclerosis, hypertension and pulmonary hypertension, in order to provide a theoretical basis for the further study of vascular remodeling.
Collapse
Affiliation(s)
- Han Xu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yu He
- Cardiovascular Surgery Department, The First Affiliated Hospital of Xi'an Jiaotong University, Xian, China
| | - Tianying Hong
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Cong Bi
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jing Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Jing Li
| | - Mingfeng Xia
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- *Correspondence: Mingfeng Xia
| |
Collapse
|
45
|
Engineering Smooth Muscle to Understand Extracellular Matrix Remodeling and Vascular Disease. Bioengineering (Basel) 2022; 9:bioengineering9090449. [PMID: 36134994 PMCID: PMC9495899 DOI: 10.3390/bioengineering9090449] [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: 08/08/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 11/29/2022] Open
Abstract
The vascular smooth muscle is vital for regulating blood pressure and maintaining cardiovascular health, and the resident smooth muscle cells (SMCs) in blood vessel walls rely on specific mechanical and biochemical signals to carry out these functions. Any slight change in their surrounding environment causes swift changes in their phenotype and secretory profile, leading to changes in the structure and functionality of vessel walls that cause pathological conditions. To adequately treat vascular diseases, it is essential to understand how SMCs crosstalk with their surrounding extracellular matrix (ECM). Here, we summarize in vivo and traditional in vitro studies of pathological vessel wall remodeling due to the SMC phenotype and, conversely, the SMC behavior in response to key ECM properties. We then analyze how three-dimensional tissue engineering approaches provide opportunities to model SMCs’ response to specific stimuli in the human body. Additionally, we review how applying biomechanical forces and biochemical stimulation, such as pulsatile fluid flow and secreted factors from other cell types, allows us to study disease mechanisms. Overall, we propose that in vitro tissue engineering of human vascular smooth muscle can facilitate a better understanding of relevant cardiovascular diseases using high throughput experiments, thus potentially leading to therapeutics or treatments to be tested in the future.
Collapse
|
46
|
Xu J, Zhong Y, Yin H, Linneman J, Luo Y, Xia S, Xia Q, Yang L, Huang X, Kang K, Wang J, Niu Y, Li L, Gou D. Methylation-mediated silencing of PTPRD induces pulmonary hypertension by promoting pulmonary arterial smooth muscle cell migration via the PDGFRB/PLCγ1 axis. J Hypertens 2022; 40:1795-1807. [PMID: 35848503 PMCID: PMC9451921 DOI: 10.1097/hjh.0000000000003220] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 05/15/2022] [Accepted: 05/15/2022] [Indexed: 12/03/2022]
Abstract
OBJECTIVE Pulmonary hypertension is a lethal disease characterized by pulmonary vascular remodeling and is mediated by abnormal proliferation and migration of pulmonary arterial smooth muscle cells (PASMCs). Platelet-derived growth factor BB (PDGF-BB) is the most potent mitogen for PASMCs and is involved in vascular remodeling in pulmonary hypertension development. Therefore, the objective of our study is to identify novel mechanisms underlying vascular remodeling in pulmonary hypertension. METHODS We explored the effects and mechanisms of PTPRD downregulation in PASMCs and PTPRD knockdown rats in pulmonary hypertension induced by hypoxia. RESULTS We demonstrated that PTPRD is dramatically downregulated in PDGF-BB-treated PASMCs, pulmonary arteries from pulmonary hypertension rats, and blood and pulmonary arteries from lung specimens of patients with hypoxic pulmonary arterial hypertension (HPAH) and idiopathic PAH (iPAH). Subsequently, we found that PTPRD was downregulated by promoter methylation via DNMT1. Moreover, we found that PTPRD knockdown altered cell morphology and migration in PASMCs via modulating focal adhesion and cell cytoskeleton. We have demonstrated that the increase in cell migration is mediated by the PDGFRB/PLCγ1 pathway. Furthermore, under hypoxic condition, we observed significant pulmonary arterial remodeling and exacerbation of pulmonary hypertension in heterozygous PTPRD knock-out rats compared with the wild-type group. We also demonstrated that HET group treated with chronic hypoxia have higher expression and activity of PLCγ1 in the pulmonary arteries compared with wild-type group. CONCLUSION We propose that PTPRD likely plays an important role in the process of pulmonary vascular remodeling and development of pulmonary hypertension in vivo .
Collapse
Affiliation(s)
- Junhua Xu
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong, China
| | - Yanfeng Zhong
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center
| | - Haoyang Yin
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center
| | - John Linneman
- Washington University School of Medicine, St. Louis, Missouri, USA
| | - Yixuan Luo
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center
| | - Sijian Xia
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center
| | - Qinyi Xia
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center
| | - Lei Yang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center
| | - Xingtao Huang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center
| | - Kang Kang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center
| | - Jun Wang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center
| | - Yanqin Niu
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center
| | - Li Li
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center
| | - Deming Gou
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center
| |
Collapse
|
47
|
Luo C, Li B, Liu C, Dong R, Hu C, Liu J, Hu L, Liao X, Zhou J, Xu L, Liu S, Yuan D, Jiang W, Yan J, Li Y. Lysyl oxidase family gene polymorphisms and risk of aneurysmal subarachnoid hemorrhage: a case-control study. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:925. [PMID: 36172092 PMCID: PMC9511205 DOI: 10.21037/atm-22-3484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/08/2022] [Indexed: 11/15/2022]
Abstract
Background Aneurysmal subarachnoid hemorrhage (aSAH) is a devastating disease caused by intracranial aneurysm (IA) rupture. Lysyl oxidase (LOX) family genes (LOX-like [LOXL] 1-4) have roles in collagen cross-linking in the extracellular matrix (ECM) and may be associated with IA rupture. We aimed to explore the association between LOX polymorphisms and the risk of aSAH. Methods This case-control study included 2 cohorts: 133 single ruptured and 115 unruptured IA patients, and 65 multiple ruptured and 71 unruptured IA patients. Genotyping of 27 single nucleotide polymorphisms (SNPs) in LOX was performed. Logistic regression analysis was performed to calculate the odds ratios (ORs) and 95% confidence intervals (CIs) of the SNPs of LOX and the risk of aSAH. Results LOX rs1800449 and LOXL4 rs3793692 were positively associated with the risk of single IA rupture in the recessive model (OR =5.66, 2.06; 95% CI =1.22–26.24, 1.11–3.82, respectively) and LOX rs10519694 demonstrated a protective effect on single IA rupture (dominant model: OR =0.42, 95% CI =0.21–0.83; recessive model: OR =0.16, 95% CI =0.04–0.65; additive model: OR =0.46, 95% CI =0.28–0.78). LOXL1 rs2165241, LOXL2 rs1063582, and LOXL3 rs17010021 showed risk effects on multiple IAs rupture. LOXL3 rs17010022 showed a protective effect on multiple IAs ruptures (dominant model: OR =0.41, 95% CI =0.21–0.82; additive model: OR =0.51, 95% CI =0.30–0.85). Conclusions LOX and LOXL4 may be susceptibility genes for single IA rupture, whereas LOXL1-3 may have a role in susceptibility to multiple IAs ruptures in the Chinese population, suggesting that LOX family genes may be associated with aSAH.
Collapse
Affiliation(s)
- Chun Luo
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Bingyang Li
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China.,Department of Information Statistics, Changsha Hospital of Traditional Chinese Medicine (Changsha Eighth Hospital), Changsha, China
| | - Chao Liu
- Department of Neurosurgery, Central Hospital of Zhuzhou, Zhuzhou, China
| | - Rui Dong
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Chongyu Hu
- Department of Neurology, Hunan People's Hospital, Changsha, China
| | - Junyu Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Liming Hu
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Xin Liao
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Macau, China
| | - Jilin Zhou
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Lu Xu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Songlin Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Dun Yuan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Weixi Jiang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Junxia Yan
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China.,The Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, China
| | - Yifeng Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
48
|
Zhao H, Gong S, Shi Y, Luo C, Qiu H, He J, Sun Y, Huang Y, Wang S, Miao Y, Wu W. The role of prolactin/vasoinhibins in cardiovascular diseases. Animal Model Exp Med 2022; 6:81-91. [PMID: 35923071 PMCID: PMC10158951 DOI: 10.1002/ame2.12264] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 07/07/2022] [Indexed: 11/12/2022] Open
Abstract
Prolactin (PRL) is a polypeptide hormone that is mainly synthesized and secreted by the lactotroph cells of the pituitary. There are two main isoforms of PRL: 23-kDa PRL (named full-length PRL) and vasoinhibins (including 5.6-18 kDa fragments). Both act as circulating hormones and cytokines to stimulate or inhibit vascular formation at different stages and neovascularization, including endothelial cell proliferation and migration, protease production, and apoptosis. However, their effects on vascular function and cardiovascular diseases are different or even contrary. In addition to the structure, secretion regulation, and signal transduction of PRL/vasoinhibins, this review focuses on the pathological mechanism and clinical significance of PRL/vasoinhibins in cardiovascular diseases.
Collapse
Affiliation(s)
- Hui Zhao
- School of Materials and Chemistry & Institute of Bismuth and Rhenium, University of Shanghai for Science and Technology, Shanghai, China.,Department of Cardio-Pulmonary Circulation, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Sugang Gong
- Department of Cardio-Pulmonary Circulation, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Yongcong Shi
- Respiratory Medicine, Dongchuan District People's Hospital, Kunming, China
| | - Cijun Luo
- Department of Cardio-Pulmonary Circulation, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Hongling Qiu
- Department of Cardio-Pulmonary Circulation, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Jing He
- Department of Cardio-Pulmonary Circulation, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Yuanyuan Sun
- Department of Cardio-Pulmonary Circulation, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Yuxia Huang
- Department of Cardio-Pulmonary Circulation, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Shang Wang
- Department of Cardio-Pulmonary Circulation, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Yuqing Miao
- School of Materials and Chemistry & Institute of Bismuth and Rhenium, University of Shanghai for Science and Technology, Shanghai, China
| | - Wenhui Wu
- Department of Cardio-Pulmonary Circulation, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| |
Collapse
|
49
|
Simões G, Pereira T, Caseiro A. Matrix metaloproteinases in vascular pathology. Microvasc Res 2022; 143:104398. [PMID: 35671836 DOI: 10.1016/j.mvr.2022.104398] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 05/01/2022] [Accepted: 06/01/2022] [Indexed: 12/07/2022]
Abstract
Vascular diseases are the main cause of morbidity and mortality. The vascular extracellular matrix (ECM) is essential in mechanical support, also regulating the cellular behavior fundamental to vascular function and homeostasis. Vascular remodeling is an adaptive response to various physiological and pathological changes and is associated with aging and vascular diseases. The aim of this review is provide a general overview of the involvement of MMPs in the pathogenesis of vascular diseases, namely, arterial hypertension, atherosclerosis, aortic aneurysms and myocardial infarction. The change in the composition of the ECM by matrix metalloproteinases (MMPs) generates a pro-inflammatory microenvironment that modifies the phenotypes of endothelial cells and vascular smooth muscle cells. They play a central role in morphogenesis, tissue repair and remodeling in response to injury, e.g., after myocardial infarction, and in progression of diseases such as atherosclerosis. Alterations in specific MMPs could influence arterial remodeling and lead to various pathological disorders such as hypertension and aneurysm formation. MMPs are regulated by endogenous tissue inhibitors of metalloproteinases (TIMPs), and the MMP/TIMP ratio generally determines the extent of ECM protein degradation and tissue remodeling. Studies are currently focused on improving the diagnostic and prognostic value of MMPs involved in the pathogenic process, increasing their therapeutic potential, and monitoring the disease. New selective MMP inhibitors may improve the specificity of these inhibitors, target specific MMPs in relevant pathological conditions and mitigate some of the side effects.
Collapse
Affiliation(s)
- Gonçalo Simões
- Politécnico de Coimbra, ESTeSC, Ciências Biomédicas Laboratoriais, Rua 5 de Outubro, 3046-854 Coimbra, Portugal.
| | - Telmo Pereira
- LABINSAÚDE - Laboratório de Investigação em Ciências Aplicadas à Saúde, Instituto Politécnico de Coimbra, ESTeSC, Rua 5 de Outubro, 3046-854 Coimbra, Portugal; Politécnico de Coimbra, ESTeSC, Fisiologia Clínica, Rua 5 de Outubro, 3046-854 Coimbra, Portugal.
| | - Armando Caseiro
- Politécnico de Coimbra, ESTeSC, Ciências Biomédicas Laboratoriais, Rua 5 de Outubro, 3046-854 Coimbra, Portugal; LABINSAÚDE - Laboratório de Investigação em Ciências Aplicadas à Saúde, Instituto Politécnico de Coimbra, ESTeSC, Rua 5 de Outubro, 3046-854 Coimbra, Portugal; Unidade I&D Química-Física Molecular, Faculdade de Ciências e Tecnologia, Universidade de Coimbra, Coimbra, Portugal.
| |
Collapse
|
50
|
Khomtchouk BB, Lee YS, Khan ML, Sun P, Mero D, Davidson MH. Targeting the cytoskeleton and extracellular matrix in cardiovascular disease drug discovery. Expert Opin Drug Discov 2022; 17:443-460. [PMID: 35258387 PMCID: PMC9050939 DOI: 10.1080/17460441.2022.2047645] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 02/24/2022] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Currently, cardiovascular disease (CVD) drug discovery has focused primarily on addressing the inflammation and immunopathology aspects inherent to various CVD phenotypes such as cardiac fibrosis and coronary artery disease. However, recent findings suggest new biological pathways for cytoskeletal and extracellular matrix (ECM) regulation across diverse CVDs, such as the roles of matricellular proteins (e.g. tenascin-C) in regulating the cellular microenvironment. The success of anti-inflammatory drugs like colchicine, which targets microtubule polymerization, further suggests that the cardiac cytoskeleton and ECM provide prospective therapeutic opportunities. AREAS COVERED Potential therapeutic targets include proteins such as gelsolin and calponin 2, which play pivotal roles in plaque development. This review focuses on the dynamic role that the cytoskeleton and ECM play in CVD pathophysiology, highlighting how novel target discovery in cytoskeletal and ECM-related genes may enable therapeutics development to alter the regulation of cellular architecture in plaque formation and rupture, cardiac contractility, and other molecular mechanisms. EXPERT OPINION Further research into the cardiac cytoskeleton and its associated ECM proteins is an area ripe for novel target discovery. Furthermore, the structural connection between the cytoskeleton and the ECM provides an opportunity to evaluate both entities as sources of potential therapeutic targets for CVDs.
Collapse
Affiliation(s)
- Bohdan B. Khomtchouk
- University of Chicago, Department of Medicine, Section of Computational Biomedicine and Biomedical Data Science, Institute for Genomics and Systems Biology, Chicago, IL USA
| | - Yoon Seo Lee
- The College of the University of Chicago, Chicago, IL USA
| | - Maha L. Khan
- The College of the University of Chicago, Chicago, IL USA
| | - Patrick Sun
- The College of the University of Chicago, Chicago, IL USA
| | | | - Michael H. Davidson
- University of Chicago, Department of Medicine, Section of Cardiology, Chicago, IL USA
| |
Collapse
|