1
|
Wang KCW, James AL, Donovan GM, Noble PB. Prenatal Origins of Obstructive Airway Disease: Starting on the Wrong Trajectory? Compr Physiol 2024; 14:5729-5762. [PMID: 39699087 DOI: 10.1002/cphy.c230019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
From the results of well-performed population health studies, we now have excellent data demonstrating that deficits in adult lung function may be present early in life, possibly as a result of developmental disorders, incurring a lifelong risk of obstructive airway diseases such as asthma and chronic obstructive pulmonary disease. Suboptimal fetal development results in intrauterine growth restriction and low birth weight at term (an outcome distinct from preterm complications), which are associated with subsequent obstructive disease. Numerous prenatal exposures and disorders compromise fetal development and these are summarized herein. Various physiological, structural, and mechanical abnormalities may result from prenatal disruption, including changes to airway smooth muscle structure-function, goblet cell biology, airway stiffness, geometry of the bronchial tree, lung parenchymal structure and mechanics, respiratory skeletal muscle contraction, and pulmonary inflammation. The literature therefore supports the need for early life intervention to prevent or correct growth defects, which may include simple nutritional or antioxidant therapy. © 2024 American Physiological Society. Compr Physiol 14:5729-5762, 2024.
Collapse
Affiliation(s)
- Kimberley C W Wang
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia
- Telethon Kids Institute, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Alan L James
- Department of Pulmonary Physiology and Sleep Medicine, West Australian Sleep Disorders Research Institute, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
- Medical School, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Graham M Donovan
- Department of Mathematics, University of Auckland, Auckland, New Zealand
| | - Peter B Noble
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia
| |
Collapse
|
2
|
Zhai M, Lei Z, Shi Y, Shi J, Zeng Y, Gong S, Jian W, Zhuang J, Yu Q, Feinberg MW, Peng W. TEAD1-Mediated Trans-Differentiation of Vascular Smooth Muscle Cells into Fibroblast-Like Cells Contributes to the Stabilization and Repair of Disrupted Atherosclerotic Plaques. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2407408. [PMID: 39665254 DOI: 10.1002/advs.202407408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 11/24/2024] [Indexed: 12/13/2024]
Abstract
Atherosclerotic plaque rupture mainly contributes to acute coronary syndrome (ACS). Insufficient repair of these plaques leads to thrombosis and subsequent ACS. Central to this process is the modulation of vascular smooth muscle cells (VSMCs) phenotypes, emphasizing their pivotal role in atherosclerotic plaque stability and healing post-disruption. Here, an expansion of FSP1+ cells in a tandem stenosis (TS) model of atherosclerotic mice is unveiled, predominantly originating from VSMCs through single-cell RNA sequencing (scRNA-seq) analyses and VSMC lineage tracing studies. Further investigation identified TEA domain transcription factor 1 (TEAD1) as the key transcription factor driving the trans-differentiation of VSMCs into fibroblast-like cells. In vivo experiments using a TS model of plaque rupture demonstrated that TEAD1 played a crucial role in promoting plaque stability and healing post-rupture through pharmacological or TEAD1-AAV treatments. Mechanistically, it is found that TEAD1 promoted the expression of fibroblast markers through the Wnt4/β-Catenin pathway, facilitating the trans-differentiation. Thus, this study illustrated that TEAD1 played a critical role in promoting the trans-differentiation of VSMCs into fibroblast-like cells and subsequent extracellular matrix production through the Wnt4/β-Catenin pathway. Consequently, this process enhanced the healing mechanisms following plaque rupture, elucidating potential therapeutic avenues for managing atherosclerotic instability.
Collapse
Affiliation(s)
- Ming Zhai
- Department of Cardiology, School of Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China
| | - Zhijun Lei
- Department of Cardiology, School of Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China
| | - Yefei Shi
- Department of Cardiology, School of Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China
| | - Jiayun Shi
- Department of Cardiology, School of Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China
| | - Yanxi Zeng
- Department of Cardiology, School of Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China
| | - Shiyu Gong
- Department of Cardiology, School of Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China
| | - Weixia Jian
- Department of Endocrinology, School of Medicine, Xinhua Hospital, Shanghai Jiaotong University, Shanghai, 200092, China
| | - Jianhui Zhuang
- Department of Cardiology, School of Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China
| | - Qing Yu
- Department of Cardiology, School of Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China
| | - Mark W Feinberg
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Wenhui Peng
- Department of Cardiology, School of Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China
| |
Collapse
|
3
|
Winner G J, Jain S, Gupta D. Unveiling novel molecules and therapeutic targets in hypertension - A narrative review. Eur J Pharmacol 2024; 984:177053. [PMID: 39393666 DOI: 10.1016/j.ejphar.2024.177053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 09/18/2024] [Accepted: 10/07/2024] [Indexed: 10/13/2024]
Abstract
Hypertension is a prevalent non-communicable disease with serious cardiovascular complications, including heart failure, myocardial infarction, and stroke, often resulting from uncontrolled hypertension. While current treatments primarily target the renin-angiotensin-aldosterone pathway, the therapeutic response remains modest in many patients, with some developing resistant hypertension. Newer therapeutic approaches aim to address hypertension from various aspects beyond conventional drugs, including targeting central nervous system pathways, inflammatory pathways, vascular smooth muscle function, and baroreceptors. Despite these advancements, each therapy faces unique clinical and mechanistic challenges that influence its clinical translatability and long-term viability. This review explores the mechanisms of novel molecules in preclinical and clinical development, highlights potential therapeutic targets, and discusses the challenges and ethical considerations related to hypertension therapeutics and their development.
Collapse
Affiliation(s)
| | - Surbhi Jain
- Aligarh Muslim University, Uttar Pradesh, India
| | | |
Collapse
|
4
|
Zhu Q, Yu Q, Liu M, Wei Y. Effectiveness of calcium supplementation in the prevention of gestational hypertension: A systematic review and meta-analysis of randomised controlled trials. Pregnancy Hypertens 2024; 38:101174. [PMID: 39608269 DOI: 10.1016/j.preghy.2024.101174] [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: 10/25/2024] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 11/30/2024]
Abstract
BACKGROUND Gestational hypertension and related disorders, such as preeclampsia, pose significant risks to maternal and foetal health. Calcium supplementation has been proposed as a preventive measure, but its effectiveness remains debated. This review assess the impact of calcium supplementation in prevention of gestational hypertension. METHODS A comprehensive literature search was conducted across multiple databases, including Scopus, EMBASE, PubMed, Web of Science, CINAHL, Cochrane CENTRAL, from inception to July 2024. Eligibility criteria included studies involving pregnant women at risk of gestational hypertension, comparing calcium supplementation to control group, and reporting on outcomes such as preeclampsia, pregnancy-induced hypertension, HELLP syndrome, preterm birth, and maternal mortality. Data were analysed using a random-effects inverse-variance model to calculate pooled risk ratios (RRs) and assess heterogeneity using Cochran's Q and I2 statistics. Publication bias was evaluated using funnel plots and Egger's test. RESULTS 22 studies with 39,270 individuals were included. Most studies had higher risk of bias. Calcium supplementation significantly reduced the risk of preeclampsia (pooled RR = 0.606, 95 %CI: 0.483-0.761, p < 0.001) and pregnancy-induced hypertension (pooled RR = 0.870, 95 %CI: 0.759-0.996, p = 0.044). However, it showed no significant effect on HELLP syndrome, preterm birth, or mortality. Heterogeneity was moderate to substantial across outcomes, and publication bias was detected for preterm birth and pregnancy-induced hypertension. CONCLUSION Calcium supplementation during pregnancy significantly reduces the risk of preeclampsia and pregnancy-induced hypertension, highlighting its value as a cost-effective intervention for improving maternal health. These findings support integrating calcium supplementation into prenatal care strategies, particularly for populations with low dietary calcium intake.
Collapse
Affiliation(s)
- Qing Zhu
- Department of Obstetrics, Jinan Central Hospital affiliated to Shandong First Medical University, No.105, Jiefang Road, Jinan 250013, China
| | - Qin Yu
- Department of Obstetrics, Jinan Central Hospital affiliated to Shandong First Medical University, No.105, Jiefang Road, Jinan 250013, China
| | - Mengyao Liu
- Department of Obstetrics, Jinan Central Hospital affiliated to Shandong First Medical University, No.105, Jiefang Road, Jinan 250013, China
| | - Yongqing Wei
- Department of Obstetrics, Jinan Central Hospital affiliated to Shandong First Medical University, No.105, Jiefang Road, Jinan 250013, China.
| |
Collapse
|
5
|
Dogru S, Yasar E, Yesilkaya A. Effects of uric acid on oxidative stress in vascular smooth muscle cells. Biomed Rep 2024; 21:171. [PMID: 39346579 PMCID: PMC11428086 DOI: 10.3892/br.2024.1859] [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: 04/29/2024] [Accepted: 08/15/2024] [Indexed: 10/01/2024] Open
Abstract
Hyperuricemia during hypertension is associated with aberrant vascular functions and increased oxidative stress, which affects endothelial dysfunction. Nevertheless, the molecular mechanisms underlying the effects of uric acid on vascular smooth muscle cells (VSMCs) through oxidative stress remain unclear. The aim of the present study was to investigate the dose- and time-dependent effects of uric acid on oxidative stress and p53 protein expression in VSMCs. VSMCs were incubated with various concentrations of uric acid (0-50 mg/dl) for different time periods (1-24 h). Thiobarbituric acid reactive substances (TBARs), protein carbonylation and nitric oxide (NO) levels were determined using appropriate assay kits. Superoxide anion release was detected using the Görlach method. Western blotting was performed to determine the protein expression levels of p53. The findings demonstrated that the application of uric acid led to an increase in protein carbonylation and superoxide anion levels while causing a decrease in NO levels. Conversely, no significant effect was observed on TBARS levels. Additionally, it was observed that high concentrations of uric acid suppressed p53 expression at 6, 12 and 24 h. The present study provided evidence that the influence of uric acid on oxidative stress was more closely associated with time than dose; however, not all effects observed were strictly time-dependent.
Collapse
Affiliation(s)
- Segun Dogru
- Department of Medical Biochemistry, Akdeniz University Medical School, 07058 Antalya, Turkey
| | - Ekrem Yasar
- Department of Biophysics, Akdeniz University Medical School, 07058 Antalya, Turkey
- Department of Biophysics, Faculty of Medicine, Erzincan Binali Yildirim University, 24100 Erzincan, Turkey
| | - Akin Yesilkaya
- Department of Medical Biochemistry, Akdeniz University Medical School, 07058 Antalya, Turkey
| |
Collapse
|
6
|
Liu Y, Huang T, Yap NA, Lim K, Ju LA. Harnessing the power of bioprinting for the development of next-generation models of thrombosis. Bioact Mater 2024; 42:328-344. [PMID: 39295733 PMCID: PMC11408160 DOI: 10.1016/j.bioactmat.2024.08.040] [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: 05/19/2024] [Revised: 08/07/2024] [Accepted: 08/29/2024] [Indexed: 09/21/2024] Open
Abstract
Thrombosis, a leading cause of cardiovascular morbidity and mortality, involves the formation of blood clots within blood vessels. Current animal models and in vitro systems have limitations in recapitulating the complex human vasculature and hemodynamic conditions, limiting the research in understanding the mechanisms of thrombosis. Bioprinting has emerged as a promising approach to construct biomimetic vascular models that closely mimic the structural and mechanical properties of native blood vessels. This review discusses the key considerations for designing bioprinted vascular conduits for thrombosis studies, including the incorporation of key structural, biochemical and mechanical features, the selection of appropriate biomaterials and cell sources, and the challenges and future directions in the field. The advancements in bioprinting techniques, such as multi-material bioprinting and microfluidic integration, have enabled the development of physiologically relevant models of thrombosis. The future of bioprinted models of thrombosis lies in the integration of patient-specific data, real-time monitoring technologies, and advanced microfluidic platforms, paving the way for personalized medicine and targeted interventions. As the field of bioprinting continues to evolve, these advanced vascular models are expected to play an increasingly important role in unraveling the complexities of thrombosis and improving patient outcomes. The continued advancements in bioprinting technologies and the collaboration between researchers from various disciplines hold great promise for revolutionizing the field of thrombosis research.
Collapse
Affiliation(s)
- Yanyan Liu
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Tao Huang
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Nicole Alexis Yap
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Khoon Lim
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
- School of Medical Sciences, The University of Sydney, Darlington, NSW 2008, Australia
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Lining Arnold Ju
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW, 2006, Australia
- Heart Research Institute, Camperdown, Newtown, NSW 2042, Australia
| |
Collapse
|
7
|
Albert AP, Jahan KS, Greenberg HZE, Shamsaldeen YA. Role for the PIP 2-binding protein myristoylated alanine-rich C-kinase substrate in vascular tissue: A novel therapeutic target for cardiovascular disease. J Cell Commun Signal 2024; 18:e12052. [PMID: 39691873 PMCID: PMC11647048 DOI: 10.1002/ccs3.12052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 09/05/2024] [Accepted: 09/23/2024] [Indexed: 12/19/2024] Open
Abstract
In vascular smooth muscle cells (VSMCs) and vascular endothelial cells (VECs), phosphatidylinositol 4,5-bisphosphate (PIP2) acts as a substrate for phospholipase C (PLC)- and phosphoinositol 3-kinase (PI3K)-mediated signaling pathways and an unmodified ligand at ion channels and other macromolecules, which are key processes in the regulation of cell physiological and pathological phenotypes. It is envisaged that these distinct roles of PIP2 are achieved by PIP2-binding proteins, which act as PIP2 buffers to produce discrete pools of PIP2 that permits targeted release within the cell. This review discusses evidence for the expression, cell distribution, and role of myristoylated alanine-rich C-kinase substrate (MARCKS), a PIP2-binding protein, in cellular signaling and function of VSMCs. The review indicates the possibilities for MARCKS as a therapeutic target for vascular disease involving dysfunctional cell proliferation and migration, endothelial barrier permeability, and vascular contractility such as atherosclerosis, systemic and pulmonary hypertension, and sepsis.
Collapse
Affiliation(s)
- Anthony P. Albert
- Vascular Biology SectionCardiovascular & Genomics Research InstituteCity St. George'sUniversity of LondonLondonUK
| | - Kazi S. Jahan
- Vascular Biology SectionCardiovascular & Genomics Research InstituteCity St. George'sUniversity of LondonLondonUK
| | - Harry Z. E. Greenberg
- Vascular Biology SectionCardiovascular & Genomics Research InstituteCity St. George'sUniversity of LondonLondonUK
| | | |
Collapse
|
8
|
Gaona-Tovar E, Estrada-Soto S, Ramírez-Hernández A, Arias-Durán L, Tlahuext H, Villalobos-Molina R, Almanza-Pérez JC. Vasorelaxant and tracheorelaxant effects of Bocconia arborea and their isolated benzophenanthridine alkaloids. Fitoterapia 2024; 179:106212. [PMID: 39278422 DOI: 10.1016/j.fitote.2024.106212] [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: 06/13/2024] [Revised: 08/27/2024] [Accepted: 09/10/2024] [Indexed: 09/18/2024]
Abstract
Bocconia arborea S. Watson (Papaveraceae) is an abundant medicinal plant in the North of Morelos State, Mexico, which is used for the treatment of several diseases. The aim of current investigation was to isolate the compounds responsible of the relaxant effect shown by the active extracts. Thus, phytochemical bio-guided fractionation allowed the isolation of angoline (1), dihydrosanguinarine (2), bocconarborine A (3), oxisanguinarine (4), and oxychelerithrine (5) from dichloromethanic and methanolic extracts from the bark of Bocconia arborea (Papaveraceae). The relaxant study on aortic and tracheal rat rings of all benzophenanthridines indicates that 1 was the most active compound of the entire series investigated. Angoline (1) induces its relaxant effect by a concentration-dependent manner through the calcium channel blockade in both tissues.
Collapse
Affiliation(s)
- Emmanuel Gaona-Tovar
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca 62209, Morelos, Mexico
| | - Samuel Estrada-Soto
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca 62209, Morelos, Mexico.
| | | | - Luis Arias-Durán
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca 62209, Morelos, Mexico
| | - Hugo Tlahuext
- Centro de Investigaciones Químicas, Universidad Autónoma del Estado de Morelos, Cuernavaca 62209, Morelos, Mexico
| | - Rafael Villalobos-Molina
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Estado de México, Mexico
| | - Julio C Almanza-Pérez
- Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Iztapalapa, Ciudad de México 09340, Mexico
| |
Collapse
|
9
|
Martinier I, Trichet L, Fernandes FM. Biomimetic tubular materials: from native tissues to a unifying view of new vascular, tracheal, gastrointestinal, oesophageal, and urinary grafts. Chem Soc Rev 2024. [PMID: 39606835 DOI: 10.1039/d4cs00429a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Repairing tubular tissues-the trachea, the esophagus, urinary and gastrointestinal tracts, and the circulatory system-from trauma or severe pathologies that require resection, calls for new, more effective graft materials. Currently, the relatively narrow family of materials available for these applications relies on synthetic polymers that fail to reproduce the biological and physical cues found in native tissues. Mimicking the structure and the composition of native tubular tissues to elaborate functional grafts is expected to outperform the materials currently in use, but remains one of the most challenging goals in the field of biomaterials. Despite their apparent diversity, tubular tissues share extensive compositional and structural features. Here, we assess the current state of the art through a dual layer model, reducing each tissue to an inner epithelial layer and an outer muscular layer. Based on this model, we examine the current strategies developed to mimic each layer and we underline how each fabrication method stands in providing a biomimetic material for future clinical translation. The analysis provided here, addressed to materials chemists, biomaterials engineers and clinical staff alike, sets new guidelines to foster the elaboration of new biomimetic materials for effective tubular tissue repair.
Collapse
Affiliation(s)
- Isabelle Martinier
- Laboratoire de Chimie de la Matière Condensée de Paris, Sorbonne Université, UMR 7574, Paris 75005, France.
| | - Léa Trichet
- Laboratoire de Chimie de la Matière Condensée de Paris, Sorbonne Université, UMR 7574, Paris 75005, France.
| | - Francisco M Fernandes
- Laboratoire de Chimie de la Matière Condensée de Paris, Sorbonne Université, UMR 7574, Paris 75005, France.
| |
Collapse
|
10
|
Dongdem JT, Etornam AE, Beletaa S, Alidu I, Kotey H, Wezena CA. The β 3-Adrenergic Receptor: Structure, Physiopathology of Disease, and Emerging Therapeutic Potential. Adv Pharmacol Pharm Sci 2024; 2024:2005589. [PMID: 39640497 PMCID: PMC11620816 DOI: 10.1155/2024/2005589] [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: 05/25/2024] [Accepted: 10/24/2024] [Indexed: 12/07/2024] Open
Abstract
The discovery and characterization of the signal cascades of the β-adrenergic receptors have made it possible to effectively target the receptors for drug development. β-Adrenergic receptors are a class A rhodopsin type of G protein-coupled receptors (GPCRs) that are stimulated mainly by catecholamines and therefore mediate diverse effects of the parasympathetic nervous system in eliciting "fight or flight" type responses. They are detectable in several human tissues where they control a plethora of physiological processes and therefore contribute to the pathogenesis of several disease conditions. Given the relevance of the β-adrenergic receptor as a molecular target for many pathological conditions, this comprehensive review aims at providing an in-depth exploration of the recent advancements in β3-adrenergic receptor research. More importantly, we delve into the prospects of the β3-adrenergic receptor as a therapeutic target across a variety of clinical domains.
Collapse
Affiliation(s)
- Julius T. Dongdem
- Department of Chemical Pathology, School of Medicine, University for Development Studies, Tamale, Northern Region, Ghana
- Department of Biochemistry and Molecular Medicine, School of Medicine, University for Development Studies, Tamale, Northern Region, Ghana
| | - Axandrah E. Etornam
- Department of Biochemistry and Molecular Medicine, School of Medicine, University for Development Studies, Tamale, Northern Region, Ghana
| | - Solomon Beletaa
- Department of Biochemistry and Molecular Medicine, School of Medicine, University for Development Studies, Tamale, Northern Region, Ghana
| | - Issah Alidu
- Department of Biochemistry and Molecular Medicine, School of Medicine, University for Development Studies, Tamale, Northern Region, Ghana
| | - Hassan Kotey
- Department of Biochemistry and Molecular Medicine, School of Medicine, University for Development Studies, Tamale, Northern Region, Ghana
| | - Cletus A. Wezena
- Department of Microbiology, Faculty of Biosciences, University for Development Studies, Tamale, Northern Region, Ghana
| |
Collapse
|
11
|
Ong CW, Tan B, Hussain S, Chuangsuwanich T, Braeu FA, Cui F. Evaluation of the effect of hemodynamic factors on retinal microcirculation by using 3D confocal image-based computational fluid dynamics. Front Bioeng Biotechnol 2024; 12:1489172. [PMID: 39664882 PMCID: PMC11631608 DOI: 10.3389/fbioe.2024.1489172] [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: 08/31/2024] [Accepted: 10/31/2024] [Indexed: 12/13/2024] Open
Abstract
Purpose To investigate local hemodynamic changes resulting from elevated intraocular pressure (IOP) in different vasculature networks using a computational fluid dynamics model based on 3D reconstructed confocal microscopic images. Methods Three-dimensional rat retinal vasculature was reconstructed from confocal microscopy images using a 3D U-Net-based labeling technique, followed by manual correction. We conducted a computational fluid dynamics (CFD) analysis on different retinal vasculature networks derived from a single rat. Various venule and arteriole pressures were applied to mimic the effects of elevated intraocular pressure (IOP), a major glaucoma risk factor. An increase in IOP typically correlates with a decrease in venous pressure. We also varied the percentage of capillary dropout, simulating the loss of blood vessels within the capillary network, by reducing the volume of the normal capillary network by 10%, 30%, and 50%. Based on the output of the CFD analysis, we calculated velocity, wall shear stress (WSS), and pressure gradient for different vasculature densities. Results Arteriolar pressure, venular pressure, and capillary dropout appear to be important factors influencing wall shear stress in the rat capillary network. Our study revealed that the pressure gradient between arterioles and venules strongly affects the local wall shear stress distribution across the 3D retinal vasculature. Specifically, under a pressure gradient of 3,250 Pa, the wall shear stress was found to vary between 0 and 20 Pa, with the highest shear stress observed in the region of the superficial layer. Additionally, capillary dropout led to a 25% increase or decrease in wall shear stress in affected areas. Conclusion The hemodynamic differences under various arteriole and venule pressures, along with different capillary dropout conditions, could help explain the development of various optic disorders, such as glaucoma, diabetic retinopathy, and retinal vein occlusion.
Collapse
Affiliation(s)
- Chi Wei Ong
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Bingyao Tan
- Department of Ocular Imaging, Singapore Eye Research Institute, Singapore, Singapore
- SERI NTU Advanced Ocular Engineering (STANCE) Program, Nanyang Technological University, Singapore, Singapore
| | - Shaista Hussain
- Institute of High Performance Computing (IHPC), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | | | - Fabian Albert Braeu
- Department of Ocular Imaging, Singapore Eye Research Institute, Singapore, Singapore
| | - Fangsen Cui
- Institute of High Performance Computing (IHPC), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore, Singapore
| |
Collapse
|
12
|
Luo X, Pang Z, Li J, Anh M, Kim BS, Gao G. Bioengineered human arterial equivalent and its applications from vascular graft to in vitro disease modeling. iScience 2024; 27:111215. [PMID: 39555400 PMCID: PMC11565542 DOI: 10.1016/j.isci.2024.111215] [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] [Indexed: 11/19/2024] Open
Abstract
Arterial disorders such as atherosclerosis, thrombosis, and aneurysm pose significant health risks, necessitating advanced interventions. Despite progress in artificial blood vessels and animal models aimed at understanding pathogenesis and developing therapies, limitations in graft functionality and species discrepancies restrict their clinical and research utility. Addressing these issues, bioengineered arterial equivalents (AEs) with enhanced vascular functions have been developed, incorporating innovative technologies that improve clinical outcomes and enhance disease progression modeling. This review offers a comprehensive overview of recent advancements in bioengineered AEs, systematically summarizing the bioengineered technologies used to construct these AEs, and discussing their implications for clinical application and pathogenesis understanding. Highlighting current breakthroughs and future perspectives, this review aims to inform and inspire ongoing research in the field, potentially transforming vascular medicine and offering new avenues for preclinical and clinical advances.
Collapse
Affiliation(s)
- Xi Luo
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Zherui Pang
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Jinhua Li
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
- School of Medical Technology, Beijing Institute of Technology, Zhengzhou Academy of Intelligent Technology, Zhengzhou 450000, China
- Beijing Institute of Technology, Zhuhai, Beijing Institute of Technology, Zhuhai 519088, China
| | - Minjun Anh
- Medical Research Institute, Pusan National University, Yangsan 50612, Republic of Korea
| | - Byoung Soo Kim
- Medical Research Institute, Pusan National University, Yangsan 50612, Republic of Korea
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
| | - Ge Gao
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
- School of Medical Technology, Beijing Institute of Technology, Zhengzhou Academy of Intelligent Technology, Zhengzhou 450000, China
| |
Collapse
|
13
|
Shan M, Qiu F, Li P, Zhang Y, Shi L. Maternal exercise represses FGF21 via SIRT1 to improve the phenotypic transformation of vascular smooth muscle in hypertensive offspring. Hypertens Res 2024:10.1038/s41440-024-01991-2. [PMID: 39543417 DOI: 10.1038/s41440-024-01991-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 10/07/2024] [Accepted: 10/25/2024] [Indexed: 11/17/2024]
Abstract
Maternal exercise during pregnancy is widely recognized as an effective means of promoting cardiovascular health in offspring. Few studies have explored how maternal exercise impacts vascular function and phenotypic switching in hypertensive offspring, despite the known involvement of vascular structural and functional remodeling in hypertension pathogenesis. Research indicates a significant relationship between elevated blood pressure and fibroblast growth factor 21 (FGF21) levels. It remains unclear whether maternal exercise during pregnancy can improve vascular function in hypertensive offspring by regulating FGF21 and its underlying mechanisms. In this study, pregnant spontaneously hypertensive rats and Wistar-Kyoto rats were randomly assigned to either a sedentary or exercise group. The exercise group underwent weightless swimming exercise from gestation day 1 (GD1) to GD20. The aim was to investigate the epigenetic modifications mediated by histone deacetylase sirtuin 1 (SIRT1) during the fetal period and the phenotypic changes in the mesenteric arteries (MAs) of hypertensive offspring. We found that maternal exercise significantly improved vascular remodeling in hypertensive offspring. Specifically, maternal exercise upregulated SIRT1 expression, which led to decreased H3K9ac (histone H3 lysine 9 acetylation) in the promoter region of the FGF21 gene. This epigenetic modification resulted in the transcriptional downregulation of FGF21 in the MAs of hypertensive fetuses. These results suggest that maternal exercise may lower blood pressure in hypertensive offspring by regulating deacetylation of the FGF21 gene promoter region through SIRT1, thereby reversing phenotypic switching and vascular structural remodeling.
Collapse
Affiliation(s)
- Meiling Shan
- Department of Exercise Physiology, Beijing Sport University, 100084, Beijing, China
- School of Physical Education, Hubei University, 430062, Wuhan, China
| | - Fang Qiu
- Department of Exercise Physiology, Beijing Sport University, 100084, Beijing, China
| | - Peng Li
- Department of Exercise Physiology, Beijing Sport University, 100084, Beijing, China
| | - Yanyan Zhang
- Department of Exercise Physiology, Beijing Sport University, 100084, Beijing, China
- Laboratory of Sports Stress and Adaptation of General Administration of Sport, Beijing University, Beijing, 100084, China
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, 100084, Beijing, China
| | - Lijun Shi
- Department of Exercise Physiology, Beijing Sport University, 100084, Beijing, China.
- Laboratory of Sports Stress and Adaptation of General Administration of Sport, Beijing University, Beijing, 100084, China.
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, 100084, Beijing, China.
| |
Collapse
|
14
|
Zhang YY, Li JZ, Xie HQ, Jin YX, Wang WT, Zhao B, Jia JM. High-resolution vasomotion analysis reveals novel arteriole physiological features and progressive modulation of cerebral vascular networks by stroke. J Cereb Blood Flow Metab 2024; 44:1330-1348. [PMID: 38820436 PMCID: PMC11542124 DOI: 10.1177/0271678x241258576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 04/22/2024] [Accepted: 05/06/2024] [Indexed: 06/02/2024]
Abstract
Spontaneous cerebral vasomotion, characterized by ∼0.1 Hz rhythmic contractility, is crucial for brain homeostasis. However, our understanding of vasomotion is limited due to a lack of high-precision analytical methods to determine single vasomotion events at basal levels. Here, we developed a novel strategy that integrates a baseline smoothing algorithm, allowing precise measurements of vasodynamics and concomitant Ca2+ dynamics in mouse cerebral vasculature imaged by two-photon microscopy. We identified several previously unrecognized vasomotion properties under different physiological and pathological conditions, especially in ischemic stroke, which is a highly harmful brain disease that results from vessel occlusion. First, the dynamic characteristics between SMCs Ca2+ and corresponding arteriolar vasomotion are correlated. Second, compared to previous diameter-based estimations, our radius-based measurements reveal anisotropic vascular movements, enabling a more precise determination of the latency between smooth muscle cell (SMC) Ca2+ activity and vasoconstriction. Third, we characterized single vasomotion event kinetics at scales of less than 4 seconds. Finally, following pathological vasoconstrictions induced by ischemic stroke, vasoactive arterioles entered an inert state and persisted despite recanalization. In summary, we developed a highly accurate technique for analyzing spontaneous vasomotion, and our data suggested a potential strategy to reduce stroke damage by promoting vasomotion recovery.
Collapse
Affiliation(s)
- Yi-Yi Zhang
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translation Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
| | - Jin-Ze Li
- Key Laboratory of Growth Regulation and Translation Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
| | - Hui-Qi Xie
- Key Laboratory of Growth Regulation and Translation Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
| | - Yu-Xiao Jin
- Key Laboratory of Growth Regulation and Translation Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
| | - Wen-Tao Wang
- Key Laboratory of Growth Regulation and Translation Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
| | - Bingrui Zhao
- Key Laboratory of Growth Regulation and Translation Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
| | - Jie-Min Jia
- Key Laboratory of Growth Regulation and Translation Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- Laboratory of Neurovascular Biology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
| |
Collapse
|
15
|
Radom M, Rybarczyk A, Piekarz I, Formanowicz P. Algorithms for evaluation of minimal cut sets. J Biomed Inform 2024; 159:104740. [PMID: 39454703 DOI: 10.1016/j.jbi.2024.104740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/02/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024]
Abstract
OBJECTIVE We propose a way to enhance the evaluation of minimal cut sets (MCSs) in biological systems modeled by Petri nets, by providing criteria and methodology for determining their optimality in disabling specific processes without affecting critical system components. METHODS This study concerns Petri nets to model biological systems and utilizes two primary approaches for MCS evaluation. First is the analyzing impact on t-invariants to identify structural dependencies. Second is assessing the impact on potentially starved transitions caused by the inactivity of specific MCSs. This approach deal with net dynamics. These methodologies aim to offer practical tools for assessing the quality and effectiveness of MCSs. RESULTS The proposed methodologies were applied to two case studies. In the first case, a cholesterol metabolism network was analyzed to investigate how local inflammation and oxidative stress, in conjunction with cholesterol imbalances, influence the progression of atherosclerosis. The MCSs were ranked, with the top sets presented, focusing on those that disabled the fewest number of t-invariants. In the second case, a carbohydrate metabolism disorder model was examined to understand its impact on atherosclerosis progression. The analysis aimed to identify MCSs that could inhibit the atherosclerosis process by targeting specific transitions. Both studies utilized the Holmes software for calculations, demonstrating the effectiveness of the proposed evaluation methodologies in ranking MCSs for practical biological applications. CONCLUSION The algorithms proposed in this paper offer an analytical approach for evaluating the quality of MCSs in biological systems. By providing criteria for MCS optimality, these approaches have potential to enhance the utility of MCS analysis in systems biology, aiding in the understanding and manipulation of complex biological networks. Algorithm are implemented within Holmes software, an open-source project available at https://github.com/bszawulak/HolmesPN.
Collapse
Affiliation(s)
- Marcin Radom
- Institute of Computing Science, Poznan University of Technology, Piotrowo 2, Poznan, 60-965, Poland; Institute of Bioorganic Chemistry, Polish Academy of Sciences, Z. Noskowskiego 12/14, Poznan, 61-704, Poland.
| | - Agnieszka Rybarczyk
- Institute of Computing Science, Poznan University of Technology, Piotrowo 2, Poznan, 60-965, Poland; Institute of Bioorganic Chemistry, Polish Academy of Sciences, Z. Noskowskiego 12/14, Poznan, 61-704, Poland; Faculty of Electrical Engineering, Gdynia Maritime University, Gdynia, 81-225, Poland
| | - Igor Piekarz
- Institute of Computing Science, Poznan University of Technology, Piotrowo 2, Poznan, 60-965, Poland
| | - Piotr Formanowicz
- Institute of Computing Science, Poznan University of Technology, Piotrowo 2, Poznan, 60-965, Poland
| |
Collapse
|
16
|
Noh SG, Kim HW, Kim S, Chung KW, Jung YS, Yoon JH, Yu BP, Lee J, Chung HY. Senoinflammation as the underlying mechanism of aging and its modulation by calorie restriction. Ageing Res Rev 2024; 101:102503. [PMID: 39284417 DOI: 10.1016/j.arr.2024.102503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/25/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024]
Abstract
Senoinflammation is characterized by an unresolved low-grade inflammatory process that affects multiple organs and systemic functions. This review begins with a brief overview of the fundamental concepts and frameworks of senoinflammation. It is widely involved in the aging of various organs and ultimately leads to progressive systemic degeneration. Senoinflammation underlying age-related inflammation, is causally related to metabolic dysregulation and the formation of senescence-associated secretory phenotype (SASP) during aging and age-related diseases. This review discusses the biochemical evidence and molecular biology data supporting the concept of senoinflammation and its regulatory processes, highlighting the anti-aging and anti-inflammatory effects of calorie restriction (CR). Experimental data from CR studies demonstrated effective suppression of various pro-inflammatory cytokines and chemokines, lipid accumulation, and SASP during aging. In conclusion, senoinflammation represents the basic mechanism that creates a microenvironment conducive to aging and age-related diseases. Furthermore, it serves as a potential therapeutic target for mitigating aging and age-related diseases.
Collapse
Affiliation(s)
- Sang Gyun Noh
- Research Institute for Drug Development, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Hyun Woo Kim
- Research Institute for Drug Development, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Seungwoo Kim
- Department of Pharmacy, College of Pharmacy, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Ki Wung Chung
- Research Institute for Drug Development, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea; Department of Pharmacy, College of Pharmacy, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Young-Suk Jung
- Research Institute for Drug Development, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea; Department of Pharmacy, College of Pharmacy, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Jeong-Hyun Yoon
- Research Institute for Drug Development, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea; Department of Pharmacy, College of Pharmacy, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Byung Pal Yu
- Department of Physiology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Jaewon Lee
- Research Institute for Drug Development, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea; Department of Pharmacy, College of Pharmacy, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea.
| | - Hae Young Chung
- Research Institute for Drug Development, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea; Department of Pharmacy, College of Pharmacy, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea.
| |
Collapse
|
17
|
Mierke CT. Mechanosensory entities and functionality of endothelial cells. Front Cell Dev Biol 2024; 12:1446452. [PMID: 39507419 PMCID: PMC11538060 DOI: 10.3389/fcell.2024.1446452] [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: 06/09/2024] [Accepted: 10/04/2024] [Indexed: 11/08/2024] Open
Abstract
The endothelial cells of the blood circulation are exposed to hemodynamic forces, such as cyclic strain, hydrostatic forces, and shear stress caused by the blood fluid's frictional force. Endothelial cells perceive mechanical forces via mechanosensors and thus elicit physiological reactions such as alterations in vessel width. The mechanosensors considered comprise ion channels, structures linked to the plasma membrane, cytoskeletal spectrin scaffold, mechanoreceptors, and junctional proteins. This review focuses on endothelial mechanosensors and how they alter the vascular functions of endothelial cells. The current state of knowledge on the dysregulation of endothelial mechanosensitivity in disease is briefly presented. The interplay in mechanical perception between endothelial cells and vascular smooth muscle cells is briefly outlined. Finally, future research avenues are highlighted, which are necessary to overcome existing limitations.
Collapse
|
18
|
Guarner-Lans V, Soria-Castro E, Cano-Martínez A, Rubio-Ruiz ME, Zarco G, Carreón-Torres E, Grimaldo O, Castrejón-Téllez V, Pérez-Torres I. Rats Exposed to Excess Sucrose During a Critical Period Develop Inflammation and Express a Secretory Phenotype of Vascular Smooth Muscle Cells. Metabolites 2024; 14:555. [PMID: 39452936 PMCID: PMC11509398 DOI: 10.3390/metabo14100555] [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: 09/04/2024] [Revised: 10/04/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Neonatal rats that receive sucrose during a critical postnatal period (CP, days 12 to 28) develop hypertension by the time they reach adulthood. Inflammation might contribute to changes during this period and could be associated with variations in the vascular smooth muscle (VSMC) phenotype. OBJECTIVE We studied changes in inflammatory pathways that could underlie the expression of the secretory phenotype in the VSMC in the thoracic aorta of rats that received sucrose during CP. METHODS We analyzed histological changes in the aorta and the expression of the COX-2, TLR4, iNOS, eNOS, MMP-2 and -9, and β- and α-actin, the quantities of TNF-α, IL-6, and IL-1β using ELISA, and the levels of fatty acids using gas chromatography. RESULTS The aortic wall presented disorganization, decellularization, and wavy elastic fibers and an increase in the lumen area. The α- and β-actin expressions were decreased, while COX-2, TLR4, TNF-α, and the activity of IL-6 were increased. Oleic acid was increased in CP in comparison to the control group. CONCLUSIONS There is transient hypertension at the end of the CP that is accompanied by inflammation and a change in the phenotype of VSMC to the secretory phenotype. The inflammatory changes could act as epigenetic signals to determine the development of hypertension when animals reach adulthood.
Collapse
Affiliation(s)
- Verónica Guarner-Lans
- Department of Physiology, Instituto Nacional de Cardiología “Ignacio Chávez”, Juan Badiano 1, Sección XVI, Tlalpan, Mexico City 14080, Mexico; (V.G.-L.); (A.C.-M.); (M.E.R.-R.)
| | - Elizabeth Soria-Castro
- Department of Cardiovascular Biomedicine, Instituto Nacional de Cardiología “Ignacio Chávez”, Juan Badiano 1, Sección XVI, Tlalpan, Mexico City 14080, Mexico;
| | - Agustina Cano-Martínez
- Department of Physiology, Instituto Nacional de Cardiología “Ignacio Chávez”, Juan Badiano 1, Sección XVI, Tlalpan, Mexico City 14080, Mexico; (V.G.-L.); (A.C.-M.); (M.E.R.-R.)
| | - María Esther Rubio-Ruiz
- Department of Physiology, Instituto Nacional de Cardiología “Ignacio Chávez”, Juan Badiano 1, Sección XVI, Tlalpan, Mexico City 14080, Mexico; (V.G.-L.); (A.C.-M.); (M.E.R.-R.)
| | - Gabriela Zarco
- Department of Pharmacology, Instituto Nacional de Cardiología “Ignacio Chávez”, Juan Badiano 1, Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Elizabeth Carreón-Torres
- Department of Molecular Biology, Instituto Nacional de Cardiología “Ignacio Chávez”, Juan Badiano 1, Sección XVI, Tlalpan, Mexico City 14080, Mexico;
| | - Oscar Grimaldo
- Department of Physiology, Instituto Nacional de Cardiología “Ignacio Chávez”, Juan Badiano 1, Sección XVI, Tlalpan, Mexico City 14080, Mexico; (V.G.-L.); (A.C.-M.); (M.E.R.-R.)
| | - Vicente Castrejón-Téllez
- Department of Physiology, Instituto Nacional de Cardiología “Ignacio Chávez”, Juan Badiano 1, Sección XVI, Tlalpan, Mexico City 14080, Mexico; (V.G.-L.); (A.C.-M.); (M.E.R.-R.)
| | - Israel Pérez-Torres
- Department of Cardiovascular Biomedicine, Instituto Nacional de Cardiología “Ignacio Chávez”, Juan Badiano 1, Sección XVI, Tlalpan, Mexico City 14080, Mexico;
| |
Collapse
|
19
|
Burma JS, Bailey DM, Johnson NE, Griffiths JK, Burkart JJ, Soligon CA, Fletcher EKS, Javra RM, Debert CT, Schneider KJ, Dunn JF, Smirl JD. Physiological influences on neurovascular coupling: A systematic review of multimodal imaging approaches and recommendations for future study designs. Exp Physiol 2024. [PMID: 39392865 DOI: 10.1113/ep092060] [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: 06/27/2024] [Accepted: 09/23/2024] [Indexed: 10/13/2024]
Abstract
In this review, we have amalgamated the literature, taking a multimodal neuroimaging approach to quantify the relationship between neuronal firing and haemodynamics during a task paradigm (i.e., neurovascular coupling response), while considering confounding physiological influences. Original research articles that used concurrent neuronal and haemodynamic quantification in humans (n ≥ 10) during a task paradigm were included from PubMed, Scopus, Web of Science, EMBASE and PsychINFO. Articles published before 31 July 2023 were considered for eligibility. Rapid screening was completed by the first author. Two authors completed the title/abstract and full-text screening. Article quality was assessed using a modified version of the National Institutes of Health Quality Assessment Tool for Observational Cohort and Cross-Sectional Studies. A total of 364 articles were included following title/abstract and full-text screening. The most common combination was EEG/functional MRI (68.7%), with cognitive (48.1%) and visual (27.5%) tasks being the most common. The majority of studies displayed an absence/minimal control of blood pressure, arterial gas concentrations and/or heart rate (92.9%), and only 1.3% monitored these factors. A minority of studies restricted or collected data pertaining to caffeine (7.4%), exercise (0.8%), food (0.5%), nicotine (2.7%), the menstrual cycle (0.3%) or cardiorespiratory fitness levels (0.5%). The cerebrovasculature is sensitive to numerous factors; thus, to understand the neurovascular coupling response fully, better control for confounding physiological influences of blood pressure and respiratory metrics is imperative during study-design formulation. Moreover, further work should continue to examine sex-based differences, the influence of sex steroid hormone concentrations and cardiorespiratory fitness.
Collapse
Affiliation(s)
- Joel S Burma
- Cerebrovascular Concussion Laboratory, Faculty of Kinesiology, University of Calgary, Alberta, Canada
- Sport Injury Prevention Research Centre, Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
- Human Performance Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
- Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Damian M Bailey
- Neurovascular Research Laboratory, Faculty of Life Sciences and Education, University of South Wales, Pontypridd, UK
| | - Nathan E Johnson
- Cerebrovascular Concussion Laboratory, Faculty of Kinesiology, University of Calgary, Alberta, Canada
| | - James K Griffiths
- Cerebrovascular Concussion Laboratory, Faculty of Kinesiology, University of Calgary, Alberta, Canada
- Department of Biomedical Engineering, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Josh J Burkart
- Cerebrovascular Concussion Laboratory, Faculty of Kinesiology, University of Calgary, Alberta, Canada
| | - Clara A Soligon
- Sport Injury Prevention Research Centre, Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
| | - Elizabeth K S Fletcher
- Cerebrovascular Concussion Laboratory, Faculty of Kinesiology, University of Calgary, Alberta, Canada
- Sport Injury Prevention Research Centre, Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
- Human Performance Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
- Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Raelyn M Javra
- Cerebrovascular Concussion Laboratory, Faculty of Kinesiology, University of Calgary, Alberta, Canada
- Sport Injury Prevention Research Centre, Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
- Human Performance Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
- Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Chantel T Debert
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Kathryn J Schneider
- Sport Injury Prevention Research Centre, Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Sport Medicine Centre, University of Calgary, Calgary, Alberta, Canada
| | - Jeff F Dunn
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jonathan D Smirl
- Cerebrovascular Concussion Laboratory, Faculty of Kinesiology, University of Calgary, Alberta, Canada
- Sport Injury Prevention Research Centre, Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
- Human Performance Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
- Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
20
|
Dada A, da Silva RDCV, Zanovello M, Moser JC, Orengo SLD, Cavichiolo MO, Bidinha ER, Boeing T, Cechinel-Filho V, de Souza P. Comparative Analysis of the Protective Effect of Naringenin on Cardiovascular Parameters of Normotensive and Hypertensive Rats Subjected to the Myocardial Infarction Model. Pharmaceuticals (Basel) 2024; 17:1324. [PMID: 39458965 PMCID: PMC11510612 DOI: 10.3390/ph17101324] [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: 08/26/2024] [Revised: 09/28/2024] [Accepted: 09/29/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Cardiovascular diseases rank as the top global cause of mortality, particularly acute myocardial infarction (MI). MI arises from the blockage of a coronary artery, which disrupts blood flow and results in tissue death. Among therapeutic approaches, bioactives from medicinal plants emerge as promising for the development of new medicines. Objectives: This study explored the effects of naringenin (NAR 100 mg/kg), a flavonoid found in citrus fruits, in normotensive (NTR) and spontaneously hypertensive (SHR) rats, both subjected to isoproterenol (ISO 85 mg/kg)-induced MI. Results: Post-treatment assessments indicated that NAR reduced blood pressure and minimized clot formation, particularly notable in the SHR group, which helps mitigate damage related to hypertension and ISO exposure. Additionally, NAR effectively restored KCl-induced contractility in the aortas of both NTR and SHR groups. NAR treatment reduced reduced glutathione (GSH) and lipid hydroperoxides (LOOH) values and recovered the activity of the antioxidant enzymes catalase (CAT) and glutathione-s-transferase (GST) in NTR groups. Moreover, myocardial damage assessed through histological analyses was reduced in groups treated with NAR. Conclusions: The results highlight significant pathophysiological differences between the groups, suggesting that NAR has protective potential against ISO-induced cardiac damage, warranting further investigation into its protective effects and mechanisms.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Priscila de Souza
- Programa de Pós-Graduação em Ciências Farmacêuticas (PPGCF), Universidade do Vale do Itajaí (Univali), Itajai 88302-901, SC, Brazil
| |
Collapse
|
21
|
Wang H, Zeng P, Zhu PH, Wang ZF, Cai YJ, Deng CY, Yang H, Mai LP, Zhang MZ, Kuang SJ, Rao F, Xu JS. Downregulation of stromal interaction molecule-1 is implicated in the age-associated vasoconstriction dysfunction of aorta, intrarenal, and coronary arteries. Eur J Pharmacol 2024; 979:176832. [PMID: 39038639 DOI: 10.1016/j.ejphar.2024.176832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/28/2024] [Accepted: 07/18/2024] [Indexed: 07/24/2024]
Abstract
The contractile function of vascular smooth muscle cells (VSMCs) typically undergoes significant changes with advancing age, leading to severe vascular aging-related diseases. The precise role and mechanism of stromal interaction molecule-1 (STIM1) in age-mediated Ca2+ signaling and vasocontraction remain unclear. The connection between STIM1 and age-related vascular dysfunction was investigated using a multi-myograph system, immunohistochemical analysis, protein blotting, and SA-β-gal staining. Results showed that vasoconstrictor responses in the thoracic aorta, intrarenal artery, and coronary artery decreased with age. STIM1 knockdown in the intrarenal and coronary arteries reduced vascular tone in young mice, while no change was observed in the thoracic aorta. A significant reduction in vascular tone occurred in the STIM1 knockout group with nifedipine. In the thoracic aorta, vasoconstriction significantly decreased with age following the use of nifedipine and thapsigargin and almost disappeared after STIM1 knockdown. The proportion of senescent VSMCs increased significantly in aged mice and further increased in sm-STIM1 KO aged mice. Moreover, the expression of senescence markers p21, p16, and IL-6 significantly increased with age, with p21 expression further increased in the STIM1 knockdown aged group, but not p16 or IL-6. These findings indicate that different arteries exhibit distinct organ-specific features and that STIM1 downregulation may contribute to age-related vasoconstrictive dysfunction through activation of the p21 pathway.
Collapse
Affiliation(s)
- Hao Wang
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Peng Zeng
- Medical Research Institute, Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Peng-Hao Zhu
- The First Clinical College, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Zi-Fan Wang
- Medical Research Institute, Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Yong-Jiang Cai
- Medical Research Institute, Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Chun-Yu Deng
- Medical Research Institute, Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Hui Yang
- Medical Research Institute, Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Li-Ping Mai
- Medical Research Institute, Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Meng-Zhen Zhang
- Medical Research Institute, Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Su-Juan Kuang
- Medical Research Institute, Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Fang Rao
- Medical Research Institute, Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.
| | - Jin-Song Xu
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
22
|
Salhi N, Deluyker D, Bito V, Zaid A, El Rhaffari L. In vitro biological activities of Calamintha nepeta L. aqueous extracts. J Appl Biomed 2024; 22:155-163. [PMID: 39434513 DOI: 10.32725/jab.2024.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 08/22/2024] [Indexed: 10/23/2024] Open
Abstract
AIM This study aimed to investigate the phenolic composition, antioxidant capacity, and toxicity of aqueous extracts of Calamintha nepeta L. leaves and their potential vasorelaxant effects. METHODS Aqueous extracts of Calamintha nepeta L. were prepared by three extraction methods: decoction, infusion, and maceration. The total phenolic contents of the extracts and their antioxidant properties were investigated. The toxicity was evaluated by Artemia salina lethality bioassay. The decoction extract was analyzed by HPLC for its chemical profile and was also used to evaluate the vasorelaxant effect on thoracic aortic rings isolated from healthy Sprague Dawley rats. Pre-contraction was induced by phenylephrine, followed by cumulative doses of the extract (0.001 up to 250 µg/ml). RESULTS Aqueous extracts of Calamintha nepeta L. showed noticeable radical scavenging and chelating activities. However, the decoction extract exhibited the most powerful antioxidant capacity. No toxicity was recorded for the extracts obtained by decoction and infusion. Caffeic acid, quercetin, and rosmarinic acid were the main identified compounds. Notably, the aqueous extract obtained by decoction induced significant relaxation in endothelium-intact aortic rings at lower concentrations, and at higher concentrations in denuded aortic rings. CONCLUSION This study reveals that Calamintha nepeta L. extracted with a decoction method possesses potent antioxidant capacity and has an endothelium-dependent vasorelaxant effect.
Collapse
Affiliation(s)
- Nadia Salhi
- Moulay Ismail University, Faculty of Sciences, Laboratory of BioActives Health and Environment, Meknes, Morocco
| | - Dorien Deluyker
- Biomedical Research Institute, UHasselt, Cardio & Organ Systems (COST), Agoralaan, 3590 Diepenbeek, Belgium
| | - Virginie Bito
- Biomedical Research Institute, UHasselt, Cardio & Organ Systems (COST), Agoralaan, 3590 Diepenbeek, Belgium
| | - Abdelhamid Zaid
- Moulay Ismail University, Faculty of Sciences, Laboratory of BioActives Health and Environment, Meknes, Morocco
| | - Lhoussaine El Rhaffari
- Moulay Ismail University, Faculty of Sciences, Laboratory of BioActives Health and Environment, Meknes, Morocco
| |
Collapse
|
23
|
Otani K, Uemura N, Funada H, Kodama T, Okada M, Yamawaki H. Alteration of reactivity in isolated mesenteric artery from Zucker fatty diabetes mellitus rats. J Pharmacol Sci 2024; 156:38-44. [PMID: 39068033 DOI: 10.1016/j.jphs.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 05/15/2024] [Accepted: 06/26/2024] [Indexed: 07/30/2024] Open
Abstract
Obesity and diabetes are major risk factors for cardiovascular diseases. Zucker fatty diabetes mellitus (ZFDM) rats are novel animal model of obesity and type 2 diabetes. We have recently reported that blood pressure in ZFDM-Leprfa/fa (Homo) rats was normal, while blood adrenaline level and heart rate were lower than those in control ZFDM-Leprfa/+ (Hetero) rats. Here, we compared the reactivity in isolated mesenteric artery between Hetero and Homo rats. Contraction induced by phenylephrine was increased, while relaxation induced by isoprenaline was decreased in Homo rats at 21-23 weeks old compared with those in Hetero rats. The mRNA expression for α1A but not β2 adrenoreceptor in Homo rats was increased. Nitric oxide (NO)-mediated relaxation induced by acetylcholine was decreased, while the mRNA expression for endothelial NO synthase (eNOS) was rather increased in mesenteric artery from Homo rats. These findings for the first time revealed that in Homo rats with reduced plasma adrenaline, blood pressure could be maintained by enhancing vascular contractility induced by adrenaline through the increased α1 adrenoceptor expression and the attenuated β2 adrenoceptor signaling. Additionally, NO-mediated endothelium-dependent relaxation is impaired perhaps due to eNOS dysfunction, which might also contribute to maintain the blood pressure in Homo rats.
Collapse
MESH Headings
- Animals
- Rats, Zucker
- Mesenteric Arteries/drug effects
- Mesenteric Arteries/physiopathology
- Male
- Receptors, Adrenergic, beta-2/genetics
- Receptors, Adrenergic, beta-2/metabolism
- Nitric Oxide Synthase Type III/metabolism
- Nitric Oxide Synthase Type III/genetics
- Nitric Oxide/metabolism
- Phenylephrine/pharmacology
- Disease Models, Animal
- Receptors, Adrenergic, alpha-1/genetics
- Receptors, Adrenergic, alpha-1/metabolism
- Isoproterenol/pharmacology
- Epinephrine/blood
- Epinephrine/pharmacology
- Diabetes Mellitus, Type 2/physiopathology
- Diabetes Mellitus, Type 2/metabolism
- Vasodilation/drug effects
- Acetylcholine/pharmacology
- Rats
- Obesity/metabolism
- Obesity/physiopathology
- Vasoconstriction/drug effects
- RNA, Messenger/metabolism
- RNA, Messenger/genetics
- Blood Pressure/drug effects
- In Vitro Techniques
Collapse
Affiliation(s)
- Kosuke Otani
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Higashi 23-35-1, Towada, Aomori, 034-8628, Japan.
| | - Naofumi Uemura
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Higashi 23-35-1, Towada, Aomori, 034-8628, Japan
| | - Hiroshi Funada
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Higashi 23-35-1, Towada, Aomori, 034-8628, Japan
| | - Tomoko Kodama
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Higashi 23-35-1, Towada, Aomori, 034-8628, Japan
| | - Muneyoshi Okada
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Higashi 23-35-1, Towada, Aomori, 034-8628, Japan
| | - Hideyuki Yamawaki
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Higashi 23-35-1, Towada, Aomori, 034-8628, Japan
| |
Collapse
|
24
|
Schinzari F, De Stefano A, Sica G, Mettimano M, Cardillo C, Tesauro M. Role of L-type Ca 2+-channels in the vasorelaxing response to finerenone in arteries of human visceral adipose tissue. Physiol Rep 2024; 12:e70062. [PMID: 39317676 PMCID: PMC11421947 DOI: 10.14814/phy2.70062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 09/11/2024] [Accepted: 09/11/2024] [Indexed: 09/26/2024] Open
Abstract
Inadequate blood supply to the expanding adipose tissue (AT) is involved in the unhealthy AT remodeling and cardiometabolic consequences of obesity. Because of the pathophysiological role of upregulated mineralocorticoid receptor (MR) signaling in the complications of obesity, this study tested the vasoactive properties of finerenone, a nonsteroidal MR antagonist, in arteries of human AT. Arteries isolated from the visceral AT of obese subjects were studied in a wire myograph. Finerenone resulted in a concentration-dependent relaxation of arteries precontracted with either the thromboxane-A2 analog U46619, ET-1, or high-K+ solution; the steroidal MR antagonist potassium canrenoate, by contrast, did not relax arteries contracted with either U46619 or high-K+ solution. Finerenone-induced relaxation after precontraction with U46619 was greater in the arteries of obese versus nonobese subjects. Mechanistically, the vasorelaxing response to finerenone was not influenced by preincubation with the nitric oxide synthase inhibitor L-NAME or by endothelium removal. Interestingly, finerenone, like the dihydropyridine Ca2+-channel blocker nifedipine, relaxed arteries contracted with the L-type Ca2+-channel agonist Bay K8644. In conclusion, finerenone relaxes arteries of human visceral AT, likely through antagonism of L-type Ca2+ channels. This finding identifies a novel mechanism by which finerenone may improve AT perfusion, hence protecting against the cardiometabolic complications of obesity.
Collapse
Affiliation(s)
- Francesca Schinzari
- Department of AgingFondazione Policlinico Universitario Agostino Gemelli IRCCSRomeItaly
| | | | - Giuseppe Sica
- Department of Experimental MedicineTor Vergata UniversityRomeItaly
| | - Marco Mettimano
- Department of Cardiovascular SciencesFondazione Policlinico Universitario Agostino Gemelli IRCCSRomeItaly
| | - Carmine Cardillo
- Department of Translational Medicine and SurgeryCatholic UniversityRomeItaly
- Department of NeurosciencesFondazione Policlinico Universitario Agostino Gemelli IRCCSRomeItaly
| | | |
Collapse
|
25
|
Deng M, Odhiambo WO, Qin M, To TT, Brewer GM, Kheshvadjian AR, Cheng C, Agak GW. Analysis of intracellular communication reveals consistent gene changes associated with early-stage acne skin. Cell Commun Signal 2024; 22:400. [PMID: 39143467 PMCID: PMC11325718 DOI: 10.1186/s12964-024-01725-4] [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: 05/10/2024] [Accepted: 06/23/2024] [Indexed: 08/16/2024] Open
Abstract
A comprehensive understanding of the intricate cellular and molecular changes governing the complex interactions between cells within acne lesions is currently lacking. Herein, we analyzed early papules from six subjects with active acne vulgaris, utilizing single-cell and high-resolution spatial RNA sequencing. We observed significant changes in signaling pathways across seven different cell types when comparing lesional skin samples (LSS) to healthy skin samples (HSS). Using CellChat, we constructed an atlas of signaling pathways for the HSS, identifying key signal distributions and cell-specific genes within individual clusters. Further, our comparative analysis revealed changes in 49 signaling pathways across all cell clusters in the LSS- 4 exhibited decreased activity, whereas 45 were upregulated, suggesting that acne significantly alters cellular dynamics. We identified ten molecules, including GRN, IL-13RA1 and SDC1 that were consistently altered in all donors. Subsequently, we focused on the function of GRN and IL-13RA1 in TREM2 macrophages and keratinocytes as these cells participate in inflammation and hyperkeratinization in the early stages of acne development. We evaluated their function in TREM2 macrophages and the HaCaT cell line. We found that GRN increased the expression of proinflammatory cytokines and chemokines, including IL-18, CCL5, and CXCL2 in TREM2 macrophages. Additionally, the activation of IL-13RA1 by IL-13 in HaCaT cells promoted the dysregulation of genes associated with hyperkeratinization, including KRT17, KRT16, and FLG. These findings suggest that modulating the GRN-SORT1 and IL-13-IL-13RA1 signaling pathways could be a promising approach for developing new acne treatments.
Collapse
Affiliation(s)
- Min Deng
- Division of Dermatology, David Geffen School of Medicine, University of California (UCLA), Los Angeles, CA, 90095, USA
| | - Woodvine O Odhiambo
- Division of Dermatology, David Geffen School of Medicine, University of California (UCLA), Los Angeles, CA, 90095, USA
| | - Min Qin
- Division of Dermatology, David Geffen School of Medicine, University of California (UCLA), Los Angeles, CA, 90095, USA
| | - Thao Tam To
- Division of Dermatology, David Geffen School of Medicine, University of California (UCLA), Los Angeles, CA, 90095, USA
| | - Gregory M Brewer
- Division of Dermatology, David Geffen School of Medicine, University of California (UCLA), Los Angeles, CA, 90095, USA
| | - Alexander R Kheshvadjian
- Division of Dermatology, David Geffen School of Medicine, University of California (UCLA), Los Angeles, CA, 90095, USA
| | - Carol Cheng
- Division of Dermatology, David Geffen School of Medicine, University of California (UCLA), Los Angeles, CA, 90095, USA
| | - George W Agak
- Division of Dermatology, David Geffen School of Medicine, University of California (UCLA), Los Angeles, CA, 90095, USA.
| |
Collapse
|
26
|
Chen X, Obukhov AG, Weisman GA, Seye CI. Basal ATP release signals through the P2Y 2 receptor to maintain the differentiated phenotype of vascular smooth muscle cells. Atherosclerosis 2024; 395:117613. [PMID: 38889566 PMCID: PMC11254552 DOI: 10.1016/j.atherosclerosis.2024.117613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 05/30/2024] [Accepted: 05/30/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND AND AIMS Vascular smooth muscle cell (VSMC) dedifferentiation contributes substantively to vascular disease. VSMCs spontaneously release low levels of ATP that modulate vessel contractility, but it is unclear if autocrine ATP signaling in VSMCs is critical to the maintenance of the VSMC contractile phenotype. METHODS We used pharmacological inhibitors to block ATP release in human aortic smooth muscle cells (HASMCs) for studying changes in VSMC differentiation marker gene expression. We employed RNA interference and generated mice with SMC-specific inducible deletion of the P2Y2 receptor (P2Y2R) gene to evaluate resulting phenotypic alterations. RESULTS HASMCs constitutively release low levels of ATP that when blocked results in a significant decrease in VSMC differentiation marker gene expression, including smooth muscle actin (SMA), smooth muscle myosin heavy chain (SMMHC), SM-22α and calponin. Basal release of ATP represses transcriptional activation of the Krüppel-Like Factor 4 (KFL4) thereby preventing platelet-derived growth factor-BB (PDGF-BB) from inhibiting expression of SMC contractile phenotype markers. SMC-restricted conditional deletion of P2Y2R evoked dedifferentiation characterized by decreases in aortic contractility and contractile phenotype markers expression. This loss was accompanied by a transition to the synthetic phenotype with the acquisition of extracellular matrix (ECM) proteins characteristic of dedifferentiation, such as osteopontin and vimentin. CONCLUSIONS Our data establish the first direct evidence that an autocrine ATP release mechanism maintains SMC cytoskeletal protein expression by inhibiting VSMCs from transitioning to a synthetic phenotype, and further demonstrate that activation of the P2Y2R by basally released ATP is required for maintenance of the differentiated VSMC phenotype.
Collapse
Affiliation(s)
- Xingjuan Chen
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, China; Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, 635 Barnhill Drive MS 360A, Indianapolis, IN, 46202, USA
| | - Alexander G Obukhov
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, 635 Barnhill Drive MS 360A, Indianapolis, IN, 46202, USA
| | - Gary A Weisman
- Department of Biochemistry, Christopher S. Bond Life Sciences Center, University of Missouri, 1201 Rollins Road, Columbia, MO, 65211, USA
| | - Cheikh I Seye
- Department of Biochemistry, Christopher S. Bond Life Sciences Center, University of Missouri, 1201 Rollins Road, Columbia, MO, 65211, USA.
| |
Collapse
|
27
|
de Paula VF, Tardelli LP, Amaral SL. Dexamethasone-Induced Arterial Stiffening Is Attenuated by Training due to a Better Balance Between Aortic Collagen and Elastin Levels. Cardiovasc Drugs Ther 2024; 38:693-703. [PMID: 36795192 DOI: 10.1007/s10557-023-07438-z] [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] [Accepted: 02/02/2023] [Indexed: 02/17/2023]
Abstract
PURPOSE Although the cardioprotective benefits of exercise training are well known, the effects of training on dexamethasone (DEX)-induced arterial stiffness are still unclear. This study was aimed at investigating the mechanisms induced by training to prevent DEX-induced arterial stiffness. METHODS Wistar rats were allocated into 4 groups and submitted to combined training (aerobic and resistance exercises, on alternate days, 60% of maximal capacity, for 74 d) or were kept sedentary: sedentary control rats (SC), DEX-treated sedentary rats (DS), combined training control (CT), and DEX-treated trained rats (DT). During the last 14 d, rats were treated with DEX (50 μg/kg per body weight, per day, s.c.) or saline. RESULTS DEX increased PWV (+44% vs +5% m/s, for DS vs SC, p<0.001) and increased aortic COL 3 protein level (+75%) in DS. In addition, PWV was correlated with COL3 levels (r=0.682, p<0.0001). Aortic elastin and COL1 protein levels remained unchanged. On the other hand, the trained and treated groups showed lower PWV values (-27% m/s, p<0.001) vs DS and lower values of aortic and femoral COL3 compared with DS. CONCLUSION As DEX is widely used in several situations, the clinical relevance of this study is that the maintenance of good physical capacity throughout life can be crucial to alleviate some of its side effects, such as arterial stiffness.
Collapse
Affiliation(s)
- Vinicius F de Paula
- Joint Graduate Program in Physiological Sciences, PIPGCF UFSCar/UNESP, Rodovia Washington Luiz, km 235, São Carlos, SP, 13565-905, Brazil
- Department of Physical Education, São Paulo State University (UNESP), School of Sciences, Av. Eng. Luiz Edmundo Carrijo Coube, 14-01, Bauru, SP, 17033-360, Brazil
| | - Lidieli P Tardelli
- Department of Physical Education, São Paulo State University (UNESP), School of Sciences, Av. Eng. Luiz Edmundo Carrijo Coube, 14-01, Bauru, SP, 17033-360, Brazil
| | - Sandra L Amaral
- Joint Graduate Program in Physiological Sciences, PIPGCF UFSCar/UNESP, Rodovia Washington Luiz, km 235, São Carlos, SP, 13565-905, Brazil.
- Department of Physical Education, São Paulo State University (UNESP), School of Sciences, Av. Eng. Luiz Edmundo Carrijo Coube, 14-01, Bauru, SP, 17033-360, Brazil.
| |
Collapse
|
28
|
Mohammed KAK, Madeddu P, Avolio E. MEK inhibitors: a promising targeted therapy for cardiovascular disease. Front Cardiovasc Med 2024; 11:1404253. [PMID: 39011492 PMCID: PMC11247000 DOI: 10.3389/fcvm.2024.1404253] [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: 03/20/2024] [Accepted: 06/13/2024] [Indexed: 07/17/2024] Open
Abstract
Cardiovascular disease (CVD) represents the leading cause of mortality and disability all over the world. Identifying new targeted therapeutic approaches has become a priority of biomedical research to improve patient outcomes and quality of life. The RAS-RAF-MEK (mitogen-activated protein kinase kinase)-ERK (extracellular signal-regulated kinase) pathway is gaining growing interest as a potential signaling cascade implicated in the pathogenesis of CVD. This pathway is pivotal in regulating cellular processes like proliferation, growth, migration, differentiation, and survival, which are vital in maintaining cardiovascular homeostasis. In addition, ERK signaling is involved in controlling angiogenesis, vascular tone, myocardial contractility, and oxidative stress. Dysregulation of this signaling cascade has been linked to cell dysfunction and vascular and cardiac pathological remodeling, which contribute to the onset and progression of CVD. Recent and ongoing research has provided insights into potential therapeutic interventions targeting the RAS-RAF-MEK-ERK pathway to improve cardiovascular pathologies. Preclinical studies have demonstrated the efficacy of targeted therapy with MEK inhibitors (MEKI) in attenuating ERK activation and mitigating CVD progression in animal models. In this article, we first describe how ERK signaling contributes to preserving cardiovascular health. We then summarize current knowledge of the roles played by ERK in the development and progression of cardiac and vascular disorders, including atherosclerosis, myocardial infarction, cardiac hypertrophy, heart failure, and aortic aneurysm. We finally report novel therapeutic strategies for these CVDs encompassing MEKI and discuss advantages, challenges, and future developments for MEKI therapeutics.
Collapse
Affiliation(s)
- Khaled A K Mohammed
- Bristol Heart Institute, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Department of Cardiothoracic Surgery, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Paolo Madeddu
- Bristol Heart Institute, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Elisa Avolio
- Bristol Heart Institute, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
29
|
Biswas PK, Park J. Applications, challenges, and prospects of induced pluripotent stem cells for vascular disease. Mol Cells 2024; 47:100077. [PMID: 38825189 PMCID: PMC11260847 DOI: 10.1016/j.mocell.2024.100077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/23/2024] [Accepted: 05/27/2024] [Indexed: 06/04/2024] Open
Abstract
Vascular disease, including heart disease, stroke, and peripheral arterial disease, is one of the leading causes of death and disability and represents a significant global health issue. Since the development of human induced pluripotent stem cells (hiPSCs) in 2007, hiPSCs have provided unique and tremendous opportunities for studying human pathophysiology, disease modeling, and drug discovery in the field of regenerative medicine. In this review, we discuss vascular physiology and related diseases, the current methods for generating vascular cells (eg, endothelial cells, smooth muscle cells, and pericytes) from hiPSCs, and describe the opportunities and challenges to the clinical applications of vascular organoids, tissue-engineered blood vessels, and vessels-on-a-chip. We then explore how hiPSCs can be used to study and treat inherited vascular diseases and discuss the current challenges and future prospects. In the future, it will be essential to develop vascularized organoids or tissues that can simultaneously undergo shear stress and cyclic stretching. This development will not only increase their maturity and function but also enable effective and innovative disease modeling and drug discovery.
Collapse
Affiliation(s)
- Polash Kumar Biswas
- Department of Physiology, College of Medicine, Hallym University, Chuncheon-si, Gangwon-do 24252, South Korea
| | - Jinkyu Park
- Department of Physiology, College of Medicine, Hallym University, Chuncheon-si, Gangwon-do 24252, South Korea; Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale School of Medicine, New Haven, CT 06511, USA.
| |
Collapse
|
30
|
Abbas M, Goodney G, Vargas JD, Gaye A. Transcriptome Study of 2 Black Cohorts Reveals cis Long Noncoding RNAs Associated With Hypertension-Related mRNAs. J Am Heart Assoc 2024; 13:e034417. [PMID: 38818927 PMCID: PMC11255619 DOI: 10.1161/jaha.124.034417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 05/06/2024] [Indexed: 06/01/2024]
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) have emerged as critical regulators of the expression of genes involved in cardiovascular diseases. This project aims to identify circulating lncRNAs associated with protein-coding mRNAs differentially expressed between hypertensive and normotensive individuals and establish their link with hypertension. METHODS AND RESULTS The analyses were conducted in 3 main steps: (1) an unbiased whole blood transcriptome-wide analysis was conducted to identify and replicate protein-coding genes differentially expressed by hypertension status in 497 and 179 Black individuals from the GENE-FORECAST (Genomics, Environmental Factors and the Social Determinants of Cardiovascular Disease in African-Americans Study) and MH-GRID (Minority Health Genomics and Translational Research Bio-Repository Database) studies, respectively. Subsequently, (2) proximal lncRNAs, termed cis lncRNA quantitative trait loci, associated with each mRNA were identified in the GENE-FORECAST study and replicated in the MH-GRID study. Finally, (3) the lncRNA quantitative trait loci were used as predictors in a random forest model to predict hypertension in both data sets. A total of 129 mRNAs were significantly differentially expressed between normotensive and hypertensive individuals in both data sets. The lncRNA-mRNA association analysis revealed 249 cis lncRNA quantitative trait loci associated with 102 mRNAs, including VAMP2 (vesicle-associated membrane protein 2), mitogen-activated protein kinase kinase 3, CCAAT enhancer binding protein beta, and lymphocyte antigen 6 complex, locus E. The 249 lncRNA quantitative trait loci predicted hypertension with an area under the curve of 0.79 and 0.71 in GENE-FORECAST and MH-GRID studies, respectively. CONCLUSIONS This study leveraged a significant sample of Black individuals, a population facing a disproportionate burden of hypertension. The analyses unveiled a total of 271 lncRNA-mRNA relationships involving mRNAs that play critical roles in vascular pathways relevant to blood pressure regulation. The compelling findings, consistent across 2 independent data sets, establish a reliable foundation for designing in vitro/in vivo experiments.
Collapse
Affiliation(s)
- Malak Abbas
- National Human Genome Research Institute, National Institutes of HealthBethesdaMD
| | - Gabriel Goodney
- National Human Genome Research Institute, National Institutes of HealthBethesdaMD
| | | | - Amadou Gaye
- National Human Genome Research Institute, National Institutes of HealthBethesdaMD
| |
Collapse
|
31
|
Nwokocha C, Palacios J, Ojukwu VE, Nna VU, Owu DU, Nwokocha M, McGrowder D, Orie NN. Oxidant-induced disruption of vascular K + channel function: implications for diabetic vasculopathy. Arch Physiol Biochem 2024; 130:361-372. [PMID: 35757993 DOI: 10.1080/13813455.2022.2090578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 06/07/2022] [Indexed: 11/02/2022]
Abstract
Diabetes in humans a chronic metabolic disorder characterised by hyperglycaemia, it is associated with an increased risk of cardiovascular disease, disruptions to metabolism and vascular functions. It is also linked to oxidative stress and its complications. Its role in vascular dysfunctions is generally reported without detailed impact on the molecular mechanisms. Potassium ion channel (K+ channels) are key regulators of vascular tone, and as membrane proteins, are modifiable by oxidant stress associated with diabetes. This review manuscript examined the impact of oxidant stress on vascular K+ channel functions in diabetes, its implication in vascular complications and metabolic and cardiovascular diseases.
Collapse
Affiliation(s)
| | - Javier Palacios
- Department of Pharmacy, Faculty of Health Sciences, Arturo Prat University, Iquique, Chile
| | - Victoria E Ojukwu
- Basic Medical Sciences, University of the West Indies, Mona, Kingston, Jamaica
| | - Victor Udo Nna
- Department of Physiology, College of Medical Sciences, University of Calabar, Calabar, Nigeria
| | - Daniel Udofia Owu
- Department of Physiology, College of Medical Sciences, University of Calabar, Calabar, Nigeria
| | - Magdalene Nwokocha
- Department of Pathology, Faculty of Medical Sciences, University of the West Indies, Mona, Kingston, Jamaica
| | - Donovan McGrowder
- Department of Pathology, Faculty of Medical Sciences, University of the West Indies, Mona, Kingston, Jamaica
| | - Nelson N Orie
- Centre of Metabolism and Inflammation, University College London, London, UK
| |
Collapse
|
32
|
Zhang Y, Xu Z, Shan M, Cao J, Zhou Y, Chen Y, Shi L. Arterial Smooth Muscle Cell AKAP150 Mediates Exercise-Induced Repression of Ca V1.2 Channel Function in Cerebral Arteries of Hypertensive Rats. Arterioscler Thromb Vasc Biol 2024; 44:1202-1221. [PMID: 38602101 DOI: 10.1161/atvbaha.124.319543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 03/28/2024] [Indexed: 04/12/2024]
Abstract
BACKGROUND Hypertension is a major, prevalent risk factor for the development and progression of cerebrovascular disease. Regular exercise has been recommended as an excellent choice for the large population of individuals with mild-to-moderate elevations in blood pressure, but the mechanisms that underlie its vascular-protective and antihypertensive effects remain unknown. Here, we describe a mechanism by which myocyte AKAP150 (A-kinase anchoring protein 150) inhibition induced by exercise training alleviates voltage-dependent L-type Ca2+ channel (CaV1.2) activity and restores cerebral arterial function in hypertension. METHODS Spontaneously hypertensive rats and newly generated smooth muscle-specific AKAP150 knockin mice were used to assess the role of myocyte AKAP150/CaV1.2 channel in regulating cerebral artery function after exercise intervention. RESULTS Activation of the AKAP150/PKCα (protein kinase Cα) signaling increased CaV1.2 activity and Ca2+ influx of cerebral arterial myocyte, thus enhancing vascular tone in spontaneously hypertensive rats. Smooth muscle-specific AKAP150 knockin mice were hypertensive with higher CaV1.2 channel activity and increased vascular tone. Furthermore, treatment of Ang II (angiotensin II) resulted in a more pronounced increase in blood pressure in smooth muscle-specific AKAP150 knockin mice. Exercise training significantly reduced arterial myocyte AKAP150 expression and alleviated CaV1.2 channel activity, thus restoring cerebral arterial function in spontaneously hypertensive rats and smooth muscle-specific AKAP150 knockin mice. AT1R (AT1 receptor) and AKAP150 were interacted closely in arterial myocytes. Exercise decreased the circulating Ang II and Ang II-involved AT1R-AKAP150 association in myocytes of hypertension. CONCLUSIONS The current study demonstrates that aerobic exercise ameliorates CaV1.2 channel function via inhibiting myocyte AKAP150, which contributes to reduced cerebral arterial tone in hypertension.
Collapse
MESH Headings
- Animals
- A Kinase Anchor Proteins/metabolism
- A Kinase Anchor Proteins/genetics
- Calcium Channels, L-Type/metabolism
- Calcium Channels, L-Type/genetics
- Rats, Inbred SHR
- Hypertension/physiopathology
- Hypertension/metabolism
- Hypertension/genetics
- Cerebral Arteries/metabolism
- Cerebral Arteries/physiopathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/physiopathology
- Male
- Myocytes, Smooth Muscle/metabolism
- Disease Models, Animal
- Physical Conditioning, Animal/physiology
- Protein Kinase C-alpha/metabolism
- Protein Kinase C-alpha/genetics
- Calcium Signaling
- Mice, Inbred C57BL
- Mice
- Rats
- Rats, Inbred WKY
- Angiotensin II
- Blood Pressure
- Signal Transduction
Collapse
Affiliation(s)
- Yanyan Zhang
- Department of Exercise Physiology (Y. Zhang, Z.X., M.S., J.C., Y. Zhou, Y.C., L.S.), Beijing Sport University, China
- Laboratory of Sports Stress and Adaptation of General Administration of Sport (Y. Zhang, L.S.), Beijing Sport University, China
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education (Y. Zhang, L.S.), Beijing Sport University, China
| | - Zhaoxia Xu
- Department of Exercise Physiology (Y. Zhang, Z.X., M.S., J.C., Y. Zhou, Y.C., L.S.), Beijing Sport University, China
| | - Meiling Shan
- Department of Exercise Physiology (Y. Zhang, Z.X., M.S., J.C., Y. Zhou, Y.C., L.S.), Beijing Sport University, China
| | - Jiaqi Cao
- Department of Exercise Physiology (Y. Zhang, Z.X., M.S., J.C., Y. Zhou, Y.C., L.S.), Beijing Sport University, China
| | - Yang Zhou
- Department of Exercise Physiology (Y. Zhang, Z.X., M.S., J.C., Y. Zhou, Y.C., L.S.), Beijing Sport University, China
| | - Yu Chen
- Department of Exercise Physiology (Y. Zhang, Z.X., M.S., J.C., Y. Zhou, Y.C., L.S.), Beijing Sport University, China
| | - Lijun Shi
- Department of Exercise Physiology (Y. Zhang, Z.X., M.S., J.C., Y. Zhou, Y.C., L.S.), Beijing Sport University, China
- Laboratory of Sports Stress and Adaptation of General Administration of Sport (Y. Zhang, L.S.), Beijing Sport University, China
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education (Y. Zhang, L.S.), Beijing Sport University, China
| |
Collapse
|
33
|
Deng M, Odhiambo WO, Qin M, To TT, Brewer GM, Kheshvadjian AR, Cheng C, Agak GW. Analysis of Intracellular Communication Reveals Consistent Gene Changes Associated with Early-Stage Acne Skin. RESEARCH SQUARE 2024:rs.3.rs-4402048. [PMID: 38854033 PMCID: PMC11160929 DOI: 10.21203/rs.3.rs-4402048/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
A comprehensive understanding of the intricate cellular and molecular changes governing the complex interactions between cells within acne lesions is currently lacking. Herein, we analyzed early papules from six subjects with active acne vulgaris, utilizing single-cell and high-resolution spatial RNA sequencing. We observed significant changes in signaling pathways across seven different cell types when comparing lesional skin samples (LSS) to healthy skin samples (HSS). Using CellChat, we constructed an atlas of signaling pathways for the HSS, identifying key signal distributions and cell-specific genes within individual clusters. Further, our comparative analysis revealed changes in 49 signaling pathways across all cell clusters in the LSS- 4 exhibited decreased activity, whereas 45 were upregulated, suggesting that acne significantly alters cellular dynamics. We identified ten molecules, including GRN, IL-13RA1 and SDC1 that were consistently altered in all donors. Subsequently, we focused on the function of GRN and IL-13RA1 in TREM2 macrophages and keratinocytes as these cells participate in inflammation and hyperkeratinization in the early stages of acne development. We evaluated their function in TREM2 macrophages and the HaCaT cell line. We found that GRN increased the expression of proinflammatory cytokines and chemokines, including IL-18, CCL5, and CXCL2 in TREM2 macrophages. Additionally, the activation of IL-13RA1 by IL-13 in HaCaT cells promoted the dysregulation of genes associated with hyperkeratinization, including KRT17, KRT16, and FLG. These findings suggest that modulating the GRN-SORT1 and IL-13-IL-13RA1 signaling pathways could be a promising approach for developing new acne treatments.
Collapse
Affiliation(s)
| | | | - Min Qin
- University of California (UCLA)
| | | | | | | | | | | |
Collapse
|
34
|
Han YS, Bandi R, Fogarty MJ, Sieck GC, Brozovich FV. Aging related decreases in NM myosin expression and contractility in a resistance vessel. Front Physiol 2024; 15:1411420. [PMID: 38808359 PMCID: PMC11130448 DOI: 10.3389/fphys.2024.1411420] [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: 04/02/2024] [Accepted: 05/01/2024] [Indexed: 05/30/2024] Open
Abstract
Introduction: Vasodilatation in response to NO is a fundamental response of the vasculature, and during aging, the vasculature is characterized by an increase in stiffness and decrease in sensitivity to NO mediated vasodilatation. Vascular tone is regulated by the activation of smooth muscle and nonmuscle (NM) myosin, which are regulated by the activities of myosin light chain kinase (MLCK) and MLC phosphatase. MLC phosphatase is a trimeric enzyme with a catalytic subunit, myosin targeting subunit (MYPT1) and 20 kDa subunit of unknown function. Alternative mRNA splicing produces LZ+/LZ- MYPT1 isoforms and the relative expression of LZ+/LZ- MYPT1 determines the sensitivity to NO mediated vasodilatation. This study tested the hypothesis that aging is associated with changes in LZ+ MYPT1 and NM myosin expression, which alter vascular reactivity. Methods: We determined MYPT1 and NM myosin expression, force and the sensitivity of both endothelial dependent and endothelial independent relaxation in tertiary mesenteric arteries of young (6mo) and elderly (24mo) Fischer344 rats. Results: The data demonstrate that aging is associated with a decrease in both the expression of NM myosin and force, but LZ+ MYPT expression and the sensitivity to both endothelial dependent and independent vasodilatation did not change. Further, smooth muscle cell hypertrophy increases the thickness of the medial layer of smooth muscle with aging. Discussion: The reduction of NM myosin may represent an aging associated compensatory mechanism to normalize the stiffness of resistance vessels in response to the increase in media thickness observed during aging.
Collapse
Affiliation(s)
- Young Soo Han
- Departments of Physiology and Biomedical Engineering and Cardiovascular Diseases, Mayo Clinic, Rochester, MN, United States
| | - Rishiraj Bandi
- Departments of Physiology and Biomedical Engineering and Cardiovascular Diseases, Mayo Clinic, Rochester, MN, United States
| | - Matthew J Fogarty
- Departments of Physiology and Biomedical Engineering and Cardiovascular Diseases, Mayo Clinic, Rochester, MN, United States
| | - Gary C Sieck
- Departments of Physiology and Biomedical Engineering and Cardiovascular Diseases, Mayo Clinic, Rochester, MN, United States
| | - Frank V Brozovich
- Departments of Physiology and Biomedical Engineering and Cardiovascular Diseases, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
35
|
Ramírez-Cuéllar J, Ferrari R, Sanz RT, Valverde-Santiago M, García-García J, Nacht AS, Castillo D, Le Dily F, Neguembor MV, Malatesta M, Bonnin S, Marti-Renom MA, Beato M, Vicent GP. LATS1 controls CTCF chromatin occupancy and hormonal response of 3D-grown breast cancer cells. EMBO J 2024; 43:1770-1798. [PMID: 38565950 PMCID: PMC11066098 DOI: 10.1038/s44318-024-00080-x] [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: 05/15/2023] [Revised: 02/05/2024] [Accepted: 02/27/2024] [Indexed: 04/04/2024] Open
Abstract
The cancer epigenome has been studied in cells cultured in two-dimensional (2D) monolayers, but recent studies highlight the impact of the extracellular matrix and the three-dimensional (3D) environment on multiple cellular functions. Here, we report the physical, biochemical, and genomic differences between T47D breast cancer cells cultured in 2D and as 3D spheroids. Cells within 3D spheroids exhibit a rounder nucleus with less accessible, more compacted chromatin, as well as altered expression of ~2000 genes, the majority of which become repressed. Hi-C analysis reveals that cells in 3D are enriched for regions belonging to the B compartment, have decreased chromatin-bound CTCF and increased fusion of topologically associating domains (TADs). Upregulation of the Hippo pathway in 3D spheroids results in the activation of the LATS1 kinase, which promotes phosphorylation and displacement of CTCF from DNA, thereby likely causing the observed TAD fusions. 3D cells show higher chromatin binding of progesterone receptor (PR), leading to an increase in the number of hormone-regulated genes. This effect is in part mediated by LATS1 activation, which favors cytoplasmic retention of YAP and CTCF removal.
Collapse
Affiliation(s)
- Julieta Ramírez-Cuéllar
- Center for Genomic Regulation (CRG), Barcelona Institute for Science and Technology (BIST) Barcelona, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Roberto Ferrari
- Center for Genomic Regulation (CRG), Barcelona Institute for Science and Technology (BIST) Barcelona, Barcelona, Spain
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Rosario T Sanz
- Molecular Biology Institute of Barcelona, Consejo Superior de Investigaciones Científicas (IBMB-CSIC), C/ Baldiri Reixac, 4-8, 08028, Barcelona, Spain
| | - Marta Valverde-Santiago
- Molecular Biology Institute of Barcelona, Consejo Superior de Investigaciones Científicas (IBMB-CSIC), C/ Baldiri Reixac, 4-8, 08028, Barcelona, Spain
| | - Judith García-García
- Molecular Biology Institute of Barcelona, Consejo Superior de Investigaciones Científicas (IBMB-CSIC), C/ Baldiri Reixac, 4-8, 08028, Barcelona, Spain
| | - A Silvina Nacht
- Center for Genomic Regulation (CRG), Barcelona Institute for Science and Technology (BIST) Barcelona, Barcelona, Spain
| | - David Castillo
- CNAG-CRG, Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Baldiri Reixac 4, Barcelona, 08028, Spain
| | - Francois Le Dily
- Center for Genomic Regulation (CRG), Barcelona Institute for Science and Technology (BIST) Barcelona, Barcelona, Spain
| | - Maria Victoria Neguembor
- Center for Genomic Regulation (CRG), Barcelona Institute for Science and Technology (BIST) Barcelona, Barcelona, Spain
| | - Marco Malatesta
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Sarah Bonnin
- Center for Genomic Regulation (CRG), Barcelona Institute for Science and Technology (BIST) Barcelona, Barcelona, Spain
| | - Marc A Marti-Renom
- Center for Genomic Regulation (CRG), Barcelona Institute for Science and Technology (BIST) Barcelona, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CNAG-CRG, Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Baldiri Reixac 4, Barcelona, 08028, Spain
- ICREA, Barcelona, Spain
| | - Miguel Beato
- Center for Genomic Regulation (CRG), Barcelona Institute for Science and Technology (BIST) Barcelona, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Guillermo P Vicent
- Center for Genomic Regulation (CRG), Barcelona Institute for Science and Technology (BIST) Barcelona, Barcelona, Spain.
- Molecular Biology Institute of Barcelona, Consejo Superior de Investigaciones Científicas (IBMB-CSIC), C/ Baldiri Reixac, 4-8, 08028, Barcelona, Spain.
| |
Collapse
|
36
|
Sekimoto T, Mori H, Koba S, Arai T, Matsukawa N, Sakai R, Yokota Y, Sato S, Tanaka H, Masaki R, Oishi Y, Ogura K, Arai K, Nomura K, Sakai K, Tsujita H, Kondo S, Tsukamoto S, Suzuki H, Shinke T. Assessment of Residual Vasospasm in Patients with Plaque Rupture or Plaque Erosion using Optical Coherence Tomography. J Atheroscler Thromb 2024; 31:559-571. [PMID: 37981329 PMCID: PMC11079494 DOI: 10.5551/jat.64556] [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: 08/27/2023] [Accepted: 10/05/2023] [Indexed: 11/21/2023] Open
Abstract
AIMS Coronary vasospasm is associated with acute coronary syndrome (ACS) and may persist during primary percutaneous coronary intervention (PCI). We aimed to elucidate the incidence, morphological characteristics, and prognostic impact of residual vasospasm in plaque rupture (PR) and plaque erosion (PE) lesions using optical coherence tomography (OCT). METHODS We enrolled 142 patients with ACS who underwent OCT-guided primary PCI. All patients received intracoronary vasodilators before OCT examination. Residual vasospasm was identified as intimal gathering and categorised as polygonal- or wavy- patterned depending on the luminal shape. A wavy pattern was defined as a curved intimal surface line. A polygonal pattern was defined as a lumen with multiple angles. The incidence of major cardiovascular events, defined as death, non-fatal myocardial infarction, stroke, and any revascularization, within 1-year of PCI was identified. RESULTS The prevalence of residual vasospasm in PR and PE was 15.1% (13 of 86) and 21.4% (12 of 56), respectively. Wavy pattern was the major shape of the residual vasospasm. Polygonal-patterned lumen was more frequently observed in PR than in PE (38.5 vs. 8.3 %). The polygonal-patterned lumens had significantly larger lipid arcs (257.9 vs. 78.0 °; P<0.01), and significantly smaller areas (1.27 vs. 1.88 mm2; P=0.05) than wavy-patterned lumens. Residual vasospasm had a prognostic impact on PR but not PE at 1-year of successful primary PCI. CONCLUSION Considerable proportion of ACS including both PR and PE had residual vasospasm with variable morphological feature and different prognostic impact.
Collapse
Affiliation(s)
- Teruo Sekimoto
- Division of Cardiology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
- Division of Cardiology, Department of Medicine, Showa University Fujigaoka Hospital, Kanagawa, Japan
| | - Hiroyoshi Mori
- Division of Cardiology, Department of Medicine, Showa University Fujigaoka Hospital, Kanagawa, Japan
| | - Shinji Koba
- Division of Cardiology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
- Division of General Medicine, Department of Perioperative Medicine, Showa University School of Dentistry, Tokyo, Japan
| | - Taito Arai
- Division of Cardiology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Naoki Matsukawa
- Department of Legal Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Rikuo Sakai
- Division of Cardiology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Yuya Yokota
- Division of Cardiology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Shunya Sato
- Division of Cardiology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Hideaki Tanaka
- Division of Cardiology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Ryota Masaki
- Division of Cardiology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Yosuke Oishi
- Division of Cardiology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Kunihiro Ogura
- Division of Cardiology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Ken Arai
- Division of Cardiology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Kosuke Nomura
- Division of Cardiology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Koshiro Sakai
- Division of Cardiology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Hiroaki Tsujita
- Division of Cardiology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Seita Kondo
- Division of Cardiology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Shigeto Tsukamoto
- Division of Cardiology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Hiroshi Suzuki
- Division of Cardiology, Department of Medicine, Showa University Fujigaoka Hospital, Kanagawa, Japan
| | - Toshiro Shinke
- Division of Cardiology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
37
|
Xie W, Chen S, Luo H, Kong C, Wang D. Critical gene signature and immunological characterization in peripheral vascular atherosclerosis: novel insights from mendelian randomization and transcriptomics. Front Genet 2024; 15:1361445. [PMID: 38660678 PMCID: PMC11039871 DOI: 10.3389/fgene.2024.1361445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 03/28/2024] [Indexed: 04/26/2024] Open
Abstract
Introduction Peripheral vascular atherosclerosis (PVA) is a chronic inflammatory disease characterized by lipid accumulation in blood vessel walls, leading to vessel narrowing and inadequate blood supply. However, the molecular mechanisms underlying PVA remain poorly understood. In this study, we employed a combination of Mendelian randomization (MR) analysis and integrated transcriptomics to identify the critical gene signature associated with PVA. Methods This study utilized three public datasets (GSE43292, GSE100927 and GSE28829) related to peripheral vascular atherosclerosis obtained from the Gene Expression Omnibus database. Instrumental variables (IVs) were identified through expression quantitative trait loci (eQTL) analysis, and two-sample MR analysis was performed using publicly available summary statistics. Disease critical genes were identified based on odds ratios and intersected with differentially expressed genes in the disease dataset. GSE28829 dataset was used to validate the screened disease critical genes. Functional enrichment analysis, GSEA analysis, and immune cell infiltration analysis were performed to further characterize the role of these genes in peripheral vascular atherosclerosis. Results A total of 26,152 gene-related SNPs were identified as IVs, and 242 disease-associated genes were identified through MR analysis. Ten disease critical genes (ARHGAP25, HCLS1, HVCN1, RBM47, LILRB1, PLAU, IFI44L, IL1B, IFI6, and CFL2) were significantly associated with peripheral vascular atherosclerosis. Functional enrichment analysis using KEGG pathways revealed enrichment in the NF-kappa B signaling pathway and osteoclast differentiation. Gene set enrichment analysis further demonstrated functional enrichment of these genes in processes related to vascular functions and immune system activation. Additionally, immune cell infiltration analysis showed differential ratios of B cells and mast cells between the disease and control groups. The correlations analysis highlights the intricate interplay between disease critical genes and immune cells associated with PVA. Conclusion In conclusion, this study provides new insights into the molecular mechanisms underlying PVA by identifying ten disease critical genes associated with the disease. These findings, supported by differential expression, functional enrichment, and immune system involvement, emphasize the role of these genes in vascular function and immune cell interactions in the context of PVA. These findings contribute to a better understanding of PVA pathogenesis and offer potential targets for further mechanistic exploration and therapeutic interventions.
Collapse
Affiliation(s)
- Wei Xie
- Department of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Institute of Cardiothoracic Vascular Disease, Nanjing University, Nanjing, China
| | - Shumin Chen
- Department of Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Hanqing Luo
- Department of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Institute of Cardiothoracic Vascular Disease, Nanjing University, Nanjing, China
| | - Chuiyu Kong
- Department of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Institute of Cardiothoracic Vascular Disease, Nanjing University, Nanjing, China
| | - Dongjin Wang
- Department of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Institute of Cardiothoracic Vascular Disease, Nanjing University, Nanjing, China
| |
Collapse
|
38
|
Wang X, He W, Chen H, Yang R, Su H, DiSanto ME, Zhang X. Alteration of the Expression and Functional Activities of Myosin II Isoforms in Enlarged Hyperplastic Prostates. J Pers Med 2024; 14:381. [PMID: 38673008 PMCID: PMC11051519 DOI: 10.3390/jpm14040381] [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: 02/05/2024] [Revised: 03/17/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
INTRODUCTION Benign prostatic hyperplasia (BPH) is a common pathologic process in aging men, and the contraction of the prostatic smooth muscles (SMs) in the stroma plays a vital role in this pathogenesis, leading to lower urinary tract symptoms (LUTSs). The isoforms of both the SM myosin (SMM) and non-muscle myosin (NMM) are associated with the contraction type of the prostatic SMs, but the mechanism has not been fully elucidated. METHODS We collected prostate tissues from 30 BPH patients receiving surgical treatments, and normal human prostate samples were obtained from 12 brain-dead men. A testosterone-induced (T-induced) rat model was built, and the epithelial hyperplastic prostates were harvested. Competitive RT-PCR was used to detect the expression of SMM isoforms. We investigated the contractility of human prostate strips in vitro in an organ bath. RESULTS The results regarding the comparisons of SMM isoforms varied between rat models and human samples. In comparison with T-induced rats and controls, competitive RT-PCR failed to show any statistically significant difference regarding the compositions of SMM isoforms. For human prostates samples, BPH patients expressed more SM-1 isoforms (66.8% vs. 60.0%, p < 0.001) and myosin light chain-17b (MLC17b) (35.9% vs. 28.5%, p < 0.05) when compared to young donors. There was a significant decrease in prostate myosin heavy chain (MHC) expression in BPH patients, with a 66.4% decrease in MHC at the mRNA level and a 51.2% decrease at the protein level. The upregulated expression of non-muscle myosin heavy chain-B (NMMHC-B) was 1.6-fold at the mRNA level and 2.1-fold at the protein level. The organ bath study showed that isolated prostate strips from BPH patients produced slower tonic contraction compared to normal humans. CONCLUSION In this study, we claim that in the enlarged prostates of patients undergoing surgeries, MHC expression significantly decreased compared to normal tissues, with elevated levels of SM-1, MLC17b, and NMMHC-B isoforms. Modifications in SMM and NMM might play a role in the tonic contractile properties of prostatic SMs and the development of LUTS/BPH. Understanding this mechanism might provide insights into the origins of LUTS/BPH and facilitate the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Xiao Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430071, China; (X.W.)
| | - Weixiang He
- Department of Urology, Xijing Hospital of the Fourth Military Medical University, Xi’an 710000, China
| | - Hui Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430071, China; (X.W.)
| | - Rui Yang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430071, China; (X.W.)
| | - Hongmei Su
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430071, China; (X.W.)
| | - Michael E. DiSanto
- Department of Surgery and Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA
| | - Xinhua Zhang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| |
Collapse
|
39
|
Asunción-Alvarez D, Palacios J, Ybañez-Julca RO, Rodriguez-Silva CN, Nwokocha C, Cifuentes F, Greensmith DJ. Calcium signaling in endothelial and vascular smooth muscle cells: sex differences and the influence of estrogens and androgens. Am J Physiol Heart Circ Physiol 2024; 326:H950-H970. [PMID: 38334967 DOI: 10.1152/ajpheart.00600.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 02/05/2024] [Accepted: 02/05/2024] [Indexed: 02/10/2024]
Abstract
Calcium signaling in vascular endothelial cells (ECs) and smooth muscle cells (VSMCs) is essential for the regulation of vascular tone. However, the changes to intracellular Ca2+ concentrations are often influenced by sex differences. Furthermore, a large body of evidence shows that sex hormone imbalance leads to dysregulation of Ca2+ signaling and this is a key factor in the pathogenesis of cardiovascular diseases. In this review, the effects of estrogens and androgens on vascular calcium-handling proteins are discussed, with emphasis on the associated genomic or nongenomic molecular mechanisms. The experimental models from which data were collected were also considered. The review highlights 1) in female ECs, transient receptor potential vanilloid 4 (TRPV4) and mitochondrial Ca2+ uniporter (MCU) enhance Ca2+-dependent nitric oxide (NO) generation. In males, only transient receptor potential canonical 3 (TRPC3) plays a fundamental role in this effect. 2) Female VSMCs have lower cytosolic Ca2+ levels than males due to differences in the activity and expression of stromal interaction molecule 1 (STIM1), calcium release-activated calcium modulator 1 (Orai1), calcium voltage-gated channel subunit-α1C (CaV1.2), Na+-K+-2Cl- symporter (NKCC1), and the Na+/K+-ATPase. 3) When compared with androgens, the influence of estrogens on Ca2+ homeostasis, vascular tone, and incidence of vascular disease is better documented. 4) Many studies use supraphysiological concentrations of sex hormones, which may limit the physiological relevance of outcomes. 5) Sex-dependent differences in Ca2+ signaling mean both sexes ought to be included in experimental design.
Collapse
Affiliation(s)
- Daniel Asunción-Alvarez
- Laboratorio de Bioquímica Aplicada, Química y Farmacia, Facultad de Ciencias de la Salud, Universidad Arturo Prat, Iquique, Chile
| | - Javier Palacios
- Laboratorio de Bioquímica Aplicada, Química y Farmacia, Facultad de Ciencias de la Salud, Universidad Arturo Prat, Iquique, Chile
| | - Roberto O Ybañez-Julca
- Departamento de Farmacología, Facultad de Farmacia y Bioquímica, Universidad Nacional de Trujillo, Trujillo, Perú
| | - Cristhian N Rodriguez-Silva
- Departamento de Farmacología, Facultad de Farmacia y Bioquímica, Universidad Nacional de Trujillo, Trujillo, Perú
| | - Chukwuemeka Nwokocha
- Department of Basic Medical Sciences Physiology Section, Faculty of Medical Sciences, The University of the West Indies, Kingston, Jamaica
| | - Fredi Cifuentes
- Laboratorio de Fisiología Experimental (EphyL), Instituto Antofagasta (IA), Universidad de Antofagasta, Antofagasta, Chile
| | - David J Greensmith
- Biomedical Research Centre, School of Science, Engineering and Environment, The University of Salford, Salford, United Kingdom
| |
Collapse
|
40
|
Hussain MS, Altamimi ASA, Afzal M, Almalki WH, Kazmi I, Alzarea SI, Gupta G, Shahwan M, Kukreti N, Wong LS, Kumarasamy V, Subramaniyan V. Kaempferol: Paving the path for advanced treatments in aging-related diseases. Exp Gerontol 2024; 188:112389. [PMID: 38432575 DOI: 10.1016/j.exger.2024.112389] [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: 11/16/2023] [Revised: 01/17/2024] [Accepted: 02/29/2024] [Indexed: 03/05/2024]
Abstract
Aging-related diseases (ARDs) are a major global health concern, and the development of effective therapies is urgently needed. Kaempferol, a flavonoid found in several plants, has emerged as a promising candidate for ameliorating ARDs. This comprehensive review examines Kaempferol's chemical properties, safety profile, and pharmacokinetics, and highlights its potential therapeutic utility against ARDs. Kaempferol's therapeutic potential is underpinned by its distinctive chemical structure, which confers antioxidative and anti-inflammatory properties. Kaempferol counteracts reactive oxygen species (ROS) and modulates crucial cellular pathways, thereby combating oxidative stress and inflammation, hallmarks of ARDs. Kaempferol's low toxicity and wide safety margins, as demonstrated by preclinical and clinical studies, further substantiate its therapeutic potential. Compelling evidence supports Kaempferol's substantial potential in addressing ARDs through several mechanisms, notably anti-inflammatory, antioxidant, and anti-apoptotic actions. Kaempferol exhibits a versatile neuroprotective effect by modulating various proinflammatory signaling pathways, including NF-kB, p38MAPK, AKT, and the β-catenin cascade. Additionally, it hinders the formation and aggregation of beta-amyloid protein and regulates brain-derived neurotrophic factors. In terms of its anticancer potential, kaempferol acts through diverse pathways, inducing apoptosis, arresting the cell cycle at the G2/M phase, suppressing epithelial-mesenchymal transition (EMT)-related markers, and affecting the phosphoinositide 3-kinase/protein kinase B signaling pathways. Subsequent studies should focus on refining dosage regimens, exploring innovative delivery systems, and conducting comprehensive clinical trials to translate these findings into effective therapeutic applications.
Collapse
Affiliation(s)
- Md Sadique Hussain
- School of Pharmaceutical Sciences, Jaipur National University, Jagatpura, 302017 Jaipur, Rajasthan, India
| | | | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, 72341, Sakaka, Aljouf, Saudi Arabia
| | - Gaurav Gupta
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, Ajman 346, United Arab Emirates
| | - Moyad Shahwan
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, Ajman 346, United Arab Emirates; Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman 346, United Arab Emirates
| | - Neelima Kukreti
- School of Pharmacy, Graphic Era Hill University, Dehradun 248007, India
| | - Ling Shing Wong
- Faculty of Health and Life Sciences, INTI International University, Nilai 71800, Malaysia
| | - Vinoth Kumarasamy
- Department of Parasitology and Medical Entomology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Cheras, 56000 Kuala Lumpur, Malaysia.
| | - Vetriselvan Subramaniyan
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Jalan Lagoon Selatan, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
41
|
Ducommun S, Jannig PR, Cervenka I, Murgia M, Mittenbühler MJ, Chernogubova E, Dias JM, Jude B, Correia JC, Van Vranken JG, Ocana-Santero G, Porsmyr-Palmertz M, McCann Haworth S, Martínez-Redondo V, Liu Z, Carlström M, Mann M, Lanner JT, Teixeira AI, Maegdefessel L, Spiegelman BM, Ruas JL. Mustn1 is a smooth muscle cell-secreted microprotein that modulates skeletal muscle extracellular matrix composition. Mol Metab 2024; 82:101912. [PMID: 38458566 PMCID: PMC10950823 DOI: 10.1016/j.molmet.2024.101912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/21/2024] [Accepted: 03/04/2024] [Indexed: 03/10/2024] Open
Abstract
OBJECTIVE Skeletal muscle plasticity and remodeling are critical for adapting tissue function to use, disuse, and regeneration. The aim of this study was to identify genes and molecular pathways that regulate the transition from atrophy to compensatory hypertrophy or recovery from injury. Here, we have used a mouse model of hindlimb unloading and reloading, which causes skeletal muscle atrophy, and compensatory regeneration and hypertrophy, respectively. METHODS We analyzed mouse skeletal muscle at the transition from hindlimb unloading to reloading for changes in transcriptome and extracellular fluid proteome. We then used qRT-PCR, immunohistochemistry, and bulk and single-cell RNA sequencing data to determine Mustn1 gene and protein expression, including changes in gene expression in mouse and human skeletal muscle with different challenges such as exercise and muscle injury. We generated Mustn1-deficient genetic mouse models and characterized them in vivo and ex vivo with regard to muscle function and whole-body metabolism. We isolated smooth muscle cells and functionally characterized them, and performed transcriptomics and proteomics analysis of skeletal muscle and aorta of Mustn1-deficient mice. RESULTS We show that Mustn1 (Musculoskeletal embryonic nuclear protein 1, also known as Mustang) is highly expressed in skeletal muscle during the early stages of hindlimb reloading. Mustn1 expression is transiently elevated in mouse and human skeletal muscle in response to intense exercise, resistance exercise, or injury. We find that Mustn1 expression is highest in smooth muscle-rich tissues, followed by skeletal muscle fibers. Muscle from heterozygous Mustn1-deficient mice exhibit differences in gene expression related to extracellular matrix and cell adhesion, compared to wild-type littermates. Mustn1-deficient mice have normal muscle and aorta function and whole-body glucose metabolism. We show that Mustn1 is secreted from smooth muscle cells, and that it is present in arterioles of the muscle microvasculature and in muscle extracellular fluid, particularly during the hindlimb reloading phase. Proteomics analysis of muscle from Mustn1-deficient mice confirms differences in extracellular matrix composition, and female mice display higher collagen content after chemically induced muscle injury compared to wild-type littermates. CONCLUSIONS We show that, in addition to its previously reported intracellular localization, Mustn1 is a microprotein secreted from smooth muscle cells into the muscle extracellular space. We explore its role in muscle ECM deposition and remodeling in homeostasis and upon muscle injury. The role of Mustn1 in fibrosis and immune infiltration upon muscle injury and dystrophies remains to be investigated, as does its potential for therapeutic interventions.
Collapse
Affiliation(s)
- Serge Ducommun
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Paulo R Jannig
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Igor Cervenka
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Marta Murgia
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi, 58/B, 35131 Padua, Italy; Max-Planck-Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Melanie J Mittenbühler
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Ekaterina Chernogubova
- Department of Medicine, Cardiovascular Unit, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - José M Dias
- Department of Cell and Molecular Biology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden; Nanomedicine and Spatial Biology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Baptiste Jude
- Molecular Muscle Physiology and Pathophysiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Jorge C Correia
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | | | - Gabriel Ocana-Santero
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Margareta Porsmyr-Palmertz
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Sarah McCann Haworth
- Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Vicente Martínez-Redondo
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Zhengye Liu
- Molecular Muscle Physiology and Pathophysiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Mattias Carlström
- Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Matthias Mann
- Max-Planck-Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Johanna T Lanner
- Molecular Muscle Physiology and Pathophysiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Ana I Teixeira
- Nanomedicine and Spatial Biology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Lars Maegdefessel
- Department of Medicine, Cardiovascular Unit, Karolinska Institutet, 171 77 Stockholm, Sweden; Institute of Molecular Vascular Medicine, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany; German Center for Cardiovascular Research DZHK, Partner Site Munich Heart Alliance, 10785 Berlin, Germany
| | - Bruce M Spiegelman
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Jorge L Ruas
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Pharmacology and Stanley and Judith Frankel Institute for Heart & Brain Health, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
42
|
Whitworth CP, Polacheck WJ. Vascular organs-on-chip made with patient-derived endothelial cells: technologies to transform drug discovery and disease modeling. Expert Opin Drug Discov 2024; 19:339-351. [PMID: 38117223 PMCID: PMC10922379 DOI: 10.1080/17460441.2023.2294947] [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: 06/30/2023] [Accepted: 12/11/2023] [Indexed: 12/21/2023]
Abstract
INTRODUCTION Vascular diseases impart a tremendous burden on healthcare systems in the United States and across the world. Efforts to improve therapeutic interventions are hindered by limitations of current experimental models. The integration of patient-derived cells with organ-on-chip (OoC) technology is a promising avenue for preclinical drug screening that improves upon traditional cell culture and animal models. AREAS COVERED The authors review induced pluripotent stem cells (iPSC) and blood outgrowth endothelial cells (BOEC) as two sources for patient-derived endothelial cells (EC). They summarize several studies that leverage patient-derived EC and OoC for precision disease modeling of the vasculature, with a focus on applications for drug discovery. They also highlight the utility of patient-derived EC in other translational endeavors, including ex vivo organogenesis and multi-organ-chip integration. EXPERT OPINION Precision disease modeling continues to mature in the academic space, but end-use by pharmaceutical companies is currently limited. To fully realize their transformative potential, OoC systems must balance their complexity with their ability to integrate with the highly standardized and high-throughput experimentation required for drug discovery and development.
Collapse
Affiliation(s)
- Chloe P Whitworth
- Curriculum in Genetics and Molecular Biology, 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, Chapel Hill, 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
- Joint Department of Biomedical Engineering, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
43
|
Damacena de Angelis C, Meddeb M, Chen N, Fisher SA. An antisense oligonucleotide efficiently suppresses splicing of an alternative exon in vascular smooth muscle in vivo. Am J Physiol Heart Circ Physiol 2024; 326:H860-H869. [PMID: 38276948 PMCID: PMC11221813 DOI: 10.1152/ajpheart.00745.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/21/2023] [Accepted: 01/18/2024] [Indexed: 01/27/2024]
Abstract
Targeting alternative exons for therapeutic gain has been achieved in a few instances and potentially could be applied more broadly. The myosin phosphatase (MP) enzyme is a critical hub upon which signals converge to regulate vessel tone. Alternative exon 24 of myosin phosphatase regulatory subunit (Mypt1 E24) is an ideal target as toggling between the two isoforms sets smooth muscle sensitivity to vasodilators such as nitric oxide (NO). This study aimed to develop a gene-based therapy to suppress splicing of Mypt1 E24 thereby switching MP enzyme to the NO-responsive isoform. CRISPR/Cas9 constructs were effective at editing of Mypt1 E24 in vitro; however, targeting of vascular smooth muscle in vivo with AAV9 was inefficient. In contrast, an octo-guanidine conjugated antisense oligonucleotide targeting the 5' splice site of Mypt1 E24 was highly efficient in vivo. It reduced the percent splicing inclusion of Mypt1 E24 from 80% to 10% in mesenteric arteries. The maximal and half-maximal effects occurred at 12.5 and 6.25 mg/kg, respectively. The effect persisted for at least 1 mo without toxicity. This highly effective splice-blocking antisense oligonucleotide could be developed as a novel therapy to reverse vascular dysfunction common to diseases such as hypertension and heart failure.NEW & NOTEWORTHY Alternative exon usage is a major driver of phenotypic diversity in all cell types including smooth muscle. However, the functional significance of most of the hundreds of thousands of alternative exons has not been defined, nor in most cases even tested. If their importance to vascular function were known these alternative exons could represent novel therapeutic targets. Here, we present injection of Vivo-morpholino splice-blocking antisense oligonucleotides as a simple, efficient, and cost-effective method for suppression of alternative exon usage in vascular smooth muscle in vivo.
Collapse
Affiliation(s)
| | - Mariam Meddeb
- Division of Cardiology, Department of Medicine, Baltimore, Maryland, United States
| | - Nelson Chen
- University of Maryland-Baltimore Scholars Program, Baltimore, Maryland, United States
| | - Steven A Fisher
- Division of Cardiology, Department of Medicine, Baltimore, Maryland, United States
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Baltimore Veterans Affairs Medical Center, Baltimore, Maryland, United States
| |
Collapse
|
44
|
Kobayashi R, Negoro H. Acute effects of the 4-4-8 breathing technique on arterial stiffness in healthy young men. Cardiol J 2024; 31:418-426. [PMID: 38348911 PMCID: PMC11229811 DOI: 10.5603/cj.96299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 12/01/2023] [Accepted: 12/23/2023] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND Increased arterial stiffness is a risk factor for cardiovascular disease. Slow, deep breathing decreases blood pressure related to arterial stiffness. The objective of the present study was to determine the acute effects of a single session of slow breathing on arterial stiffness, blood pressure, and cardiac autonomic function. METHODS Fifteen healthy men (20 ± 0 years) were administered (a) a slow breathing condition (12 consecutive breaths of 4 s of inhalation, 4 s of pause, and 8 s of exhalation through the nose, approximately 5 min per breath) and (b) a control, two-condition crossover design. Carotid-femoral artery pulse wave velocity (cfPWV), brachial-ankle PWV (baPWV), brachial blood pressure, high frequency (HF) and low frequency (LF) were measured at baseline, 30 min, 60 min and 24 h after respiratory control. RESULTS Brachial-ankle PWV and brachial systolic pressure on the 4-4-8 breathing trial decreased after 30 min of respiratory control compared to baseline (p < 0.05), but did not change on the CON trial. Carotid-femoral PWV on both trials was unchanged; HF on the 4-4-8 breathing trial increased (p < 0.05) and LF decreased (p < 0.05) after 30 min of respiratory control compared to baseline, but was unchanged on the CON trial. CONCLUSIONS These results suggest that slow breathing techniques may be effective in modulating autonomic function and improving arterial stiffness in healthy young adults.
Collapse
Affiliation(s)
- Ryota Kobayashi
- Department of Natural and Environmental Science, Teikyo University of Science, Tokyo, Japan.
| | - Hideyuki Negoro
- Department of Medicine, Nara Medical University, Nara, Japan
| |
Collapse
|
45
|
Zhang D, Ruan J, Peng S, Li J, Hu X, Zhang Y, Zhang T, Ge Y, Zhu Z, Xiao X, Zhu Y, Li X, Li T, Zhou L, Gao Q, Zheng G, Zhao B, Li X, Zhu Y, Wu J, Li W, Zhao J, Ge WP, Xu T, Jia JM. Synaptic-like transmission between neural axons and arteriolar smooth muscle cells drives cerebral neurovascular coupling. Nat Neurosci 2024; 27:232-248. [PMID: 38168932 PMCID: PMC10849963 DOI: 10.1038/s41593-023-01515-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 11/02/2023] [Indexed: 01/05/2024]
Abstract
Neurovascular coupling (NVC) is important for brain function and its dysfunction underlies many neuropathologies. Although cell-type specificity has been implicated in NVC, how active neural information is conveyed to the targeted arterioles in the brain remains poorly understood. Here, using two-photon focal optogenetics in the mouse cerebral cortex, we demonstrate that single glutamatergic axons dilate their innervating arterioles via synaptic-like transmission between neural-arteriolar smooth muscle cell junctions (NsMJs). The presynaptic parental-daughter bouton makes dual innervations on postsynaptic dendrites and on arteriolar smooth muscle cells (aSMCs), which express many types of neuromediator receptors, including a low level of glutamate NMDA receptor subunit 1 (Grin1). Disruption of NsMJ transmission by aSMC-specific knockout of GluN1 diminished optogenetic and whisker stimulation-caused functional hyperemia. Notably, the absence of GluN1 subunit in aSMCs reduced brain atrophy following cerebral ischemia by preventing Ca2+ overload in aSMCs during arteriolar constriction caused by the ischemia-induced spreading depolarization. Our findings reveal that NsMJ transmission drives NVC and open up a new avenue for studying stroke.
Collapse
Affiliation(s)
- Dongdong Zhang
- School of Life Sciences, Fudan University, Shanghai, China
- Key Laboratory of Growth Regulation and Translation Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- Laboratory of Neurovascular Biology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Laboratory of Neurovascular Biology, School of Life Sciences, Westlake University, Hangzhou, China
| | - Jiayu Ruan
- Key Laboratory of Growth Regulation and Translation Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- Laboratory of Neurovascular Biology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Laboratory of Neurovascular Biology, School of Life Sciences, Westlake University, Hangzhou, China
| | - Shiyu Peng
- Key Laboratory of Growth Regulation and Translation Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
| | - Jinze Li
- Key Laboratory of Growth Regulation and Translation Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- Laboratory of Neurovascular Biology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Laboratory of Neurovascular Biology, School of Life Sciences, Westlake University, Hangzhou, China
| | - Xu Hu
- Key Laboratory of Growth Regulation and Translation Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- Laboratory of Neurovascular Biology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Laboratory of Neurovascular Biology, School of Life Sciences, Westlake University, Hangzhou, China
| | - Yiyi Zhang
- Key Laboratory of Growth Regulation and Translation Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- Laboratory of Neurovascular Biology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Laboratory of Neurovascular Biology, School of Life Sciences, Westlake University, Hangzhou, China
| | - Tianrui Zhang
- Laboratory of Neurovascular Biology, School of Life Sciences, Westlake University, Hangzhou, China
| | - Yaping Ge
- Key Laboratory of Growth Regulation and Translation Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- Laboratory of Neurovascular Biology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Laboratory of Neurovascular Biology, School of Life Sciences, Westlake University, Hangzhou, China
| | - Zhu Zhu
- Key Laboratory of Growth Regulation and Translation Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- Laboratory of Neurovascular Biology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Laboratory of Neurovascular Biology, School of Life Sciences, Westlake University, Hangzhou, China
| | - Xian Xiao
- Key Laboratory of Growth Regulation and Translation Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- Laboratory of Neurovascular Biology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
| | - Yunxu Zhu
- Laboratory of Neurovascular Biology, School of Life Sciences, Westlake University, Hangzhou, China
| | - Xuzhao Li
- Key Laboratory of Growth Regulation and Translation Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- Laboratory of Neurovascular Biology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Laboratory of Neurovascular Biology, School of Life Sciences, Westlake University, Hangzhou, China
| | - Tingbo Li
- Key Laboratory of Growth Regulation and Translation Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- Laboratory of Neurovascular Biology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Laboratory of Neurovascular Biology, School of Life Sciences, Westlake University, Hangzhou, China
| | - Lili Zhou
- Key Laboratory of Growth Regulation and Translation Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- Laboratory of Neurovascular Biology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Laboratory of Neurovascular Biology, School of Life Sciences, Westlake University, Hangzhou, China
| | - Qingzhu Gao
- Laboratory of Neurovascular Biology, School of Life Sciences, Westlake University, Hangzhou, China
| | - Guoxiao Zheng
- Laboratory of Neurovascular Biology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
| | - Bingrui Zhao
- Key Laboratory of Growth Regulation and Translation Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- Laboratory of Neurovascular Biology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Laboratory of Neurovascular Biology, School of Life Sciences, Westlake University, Hangzhou, China
| | - Xiangqing Li
- College of Artificial Intelligence and Big Data for Medical Sciences, Shandong Academy of Medical Sciences, Shandong First Medical University, Jinan, China
| | - Yanming Zhu
- Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- Program in Speech and Hearing Bioscience and Technology, Harvard Medical School, Boston, MA, USA
| | - Jinsong Wu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- Brain Function Laboratory, Neurosurgical Institute of Fudan University, Shanghai, China
- Institute of Brain-Intelligence Technology, Zhangjiang Lab, Shanghai, China, Shanghai, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Wensheng Li
- Department of Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jingwei Zhao
- Department of Anatomy, Histology, and Embryology, Research Center of Systemic Medicine, School of Basic Medicine, and Department of Pathology of the Sir Run-Run Shaw Hospital, The Cryo-EM Center, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Woo-Ping Ge
- Chinese Institute for Brain Research, Beijing, Beijing, China
| | - Tian Xu
- Key Laboratory of Growth Regulation and Translation Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
| | - Jie-Min Jia
- Key Laboratory of Growth Regulation and Translation Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China.
- Laboratory of Neurovascular Biology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China.
- Laboratory of Neurovascular Biology, School of Life Sciences, Westlake University, Hangzhou, China.
| |
Collapse
|
46
|
Ali MA, Gioscia-Ryan R, Yang D, Sutton NR, Tyrrell DJ. Cardiovascular aging: spotlight on mitochondria. Am J Physiol Heart Circ Physiol 2024; 326:H317-H333. [PMID: 38038719 PMCID: PMC11219063 DOI: 10.1152/ajpheart.00632.2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/02/2023]
Abstract
Mitochondria are cellular organelles critical for ATP production and are particularly relevant to cardiovascular diseases including heart failure, atherosclerosis, ischemia-reperfusion injury, and cardiomyopathies. With advancing age, even in the absence of clinical disease, mitochondrial homeostasis becomes disrupted (e.g., redox balance, mitochondrial DNA damage, oxidative metabolism, and mitochondrial quality control). Mitochondrial dysregulation leads to the accumulation of damaged and dysfunctional mitochondria, producing excessive reactive oxygen species and perpetuating mitochondrial dysfunction. In addition, mitochondrial DNA, cardiolipin, and N-formyl peptides are potent activators of cell-intrinsic and -extrinsic inflammatory pathways. These age-related mitochondrial changes contribute to the development of cardiovascular diseases. This review covers the impact of aging on mitochondria and links these mechanisms to therapeutic implications for age-associated cardiovascular diseases.
Collapse
Affiliation(s)
- Md Akkas Ali
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Rachel Gioscia-Ryan
- Department of Anesthesiology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, United States
| | - Dongli Yang
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Nadia R Sutton
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, United States
| | - Daniel J Tyrrell
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, United States
| |
Collapse
|
47
|
Luu N, Bajpai A, Li R, Park S, Noor M, Ma X, Chen W. Aging-associated decline in vascular smooth muscle cell mechanosensation is mediated by Piezo1 channel. Aging Cell 2024; 23:e14036. [PMID: 37941511 PMCID: PMC10861209 DOI: 10.1111/acel.14036] [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: 05/11/2023] [Revised: 09/27/2023] [Accepted: 10/27/2023] [Indexed: 11/10/2023] Open
Abstract
Aging of the vasculature is associated with detrimental changes in vascular smooth muscle cell (VSMC) mechanosensitivity to extrinsic forces in their surrounding microenvironment. However, how chronological aging alters VSMCs' ability to sense and adapt to mechanical perturbations remains unexplored. Here, we show defective VSMC mechanosensation in aging measured with ultrasound tweezers-based micromechanical system, force instantaneous frequency spectrum, and transcriptome analyses. The study reveals that aged VSMCs adapt to a relatively inert mechanobiological state with altered actin cytoskeletal integrity, resulting in an impairment in their mechanosensitivity and dynamic mechanoresponse to mechanical perturbations. The aging-associated decline in mechanosensation behaviors is mediated by hyperactivity of Piezo1-dependent calcium signaling. Inhibition of Piezo1 alleviates vascular aging and partially restores the loss in dynamic contractile properties in aged cells. Altogether, our study reveals the signaling pathway underlying aging-associated aberrant mechanosensation in VSMC and identifies Piezo1 as a potential therapeutic mechanobiological target to alleviate vascular aging.
Collapse
Affiliation(s)
- Ngoc Luu
- Department of Biomedical EngineeringNew York UniversityBrooklynNew YorkUSA
| | - Apratim Bajpai
- Department of Mechanical and Aerospace EngineeringNew York UniversityBrooklynNew YorkUSA
| | - Rui Li
- Department of Mechanical and Aerospace EngineeringNew York UniversityBrooklynNew YorkUSA
| | - Seojin Park
- Department of Biomedical EngineeringNew York UniversityBrooklynNew YorkUSA
| | - Mahad Noor
- Department of Mechanical and Aerospace EngineeringNew York UniversityBrooklynNew YorkUSA
| | - Xiao Ma
- Department of Biomedical EngineeringNew York UniversityBrooklynNew YorkUSA
| | - Weiqiang Chen
- Department of Biomedical EngineeringNew York UniversityBrooklynNew YorkUSA
- Department of Mechanical and Aerospace EngineeringNew York UniversityBrooklynNew YorkUSA
- Laura and Isaac Perlmutter Cancer CenterNYU Langone HealthNew YorkUSA
| |
Collapse
|
48
|
Neisi A, Farhadi M, Cheraghian B, Dargahi A, Ahmadi M, Takdastan A, Ahmadi Angali K. Consumption of foods contaminated with heavy metals and their association with cardiovascular disease (CVD) using GAM software (cohort study). Heliyon 2024; 10:e24517. [PMID: 38288014 PMCID: PMC10823063 DOI: 10.1016/j.heliyon.2024.e24517] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 01/06/2024] [Accepted: 01/10/2024] [Indexed: 01/31/2024] Open
Abstract
Introduction Heavy metals can enter the environment and food through industrial activities, acid rain, chemical fertilizers, pesticides, and sewage. A large amount of these metals is dangerous because they tend to bio accumulate. A concern with these metals is the long-term, low-dose exposure seen in the general population. HMs can cause disorders in the cardiovascular system through various mechanisms such as the production of free radicals, DNA damage, lipid peroxidation, and oxidative stress. Material and method Food items measured in the present study included rice, bread, and vegetables. 210 participants (105 controls and 105 patients) were randomly selected for this study. The demographic information of the subjects was obtained from the Hoveyzeh Cohort Center. The relationship between heavy metals in food and cardiovascular diseases is investigated by The Generalized Additive Model (GAM). Result The results of the present study showed that when urine Cd was smoothed based on rice Cd, there was a significant correlation between urine Cd and Cd consumed in vegetables and rice. The GAM coefficient for urinary Cd excreted in case-control groups and Cd consumed in vegetables were 479.79(SE: 6.49-73.87) and 818.56(SE: 11.96-68.43), respectively, and for rice consumed, it was 0.03(SE: 0.015-2.103) and 0.04(SE: 0.017-2.338), respectively. The GAM coefficient for As consumption in vegetables and As in urine of case and control groups was 1.61 (SE: 9.48-0.16) and 22.36 (SE: 13.60-1.64), respectively. The same coefficient for rice consumption in case and control groups was 4.5 (SE: 0.62-7.22) and 10.48 (SE: 1.46-7.16), respectively. There was a very strong and significant correlation between the Sr in the urine of both groups and the Sr in the food consumed, so that the urinary Sr in the control group is excreted more than in the cardiovascular group. Conclusion GAM analysis indicates that As in vegetable and rice is more than the standard limitation value. Also, Sr and Cd in vegetables, rice, and bread were more than the standard limitation value. According to the GAM model As had a significant value in rice and vegetables indicating that As is more than the standard limitation value, therefore, it is associated with CVD.
Collapse
Affiliation(s)
- Abdolkazem Neisi
- Department of Environmental Health, School of Public Health and Air Pollution and Respiratory Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Majid Farhadi
- Student Research Committee, Department of Environmental Health Engineering, School of Public Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Bahman Cheraghian
- Department of Biostatistics and Epidemiology, School of Public Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Abdollah Dargahi
- Department of Environmental Health and Social Determinants of Health Research Center Khalkhal University of Medical Sciences, Khalkhal, Iran
| | - Mehdi Ahmadi
- Environmental Technologies Research Center, and Department of Environmental Health Engineering, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Afshin Takdastan
- Environmental Technologies Research Center, and Department of Environmental Health Engineering, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Kambiz Ahmadi Angali
- Department of Biostatistics and Epidemiology, School of Health, Social Determinants of Health Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
49
|
Zhang Y, Li N, Kobayashi S. Paxillin participates in the sphingosylphosphorylcholine-induced abnormal contraction of vascular smooth muscle by regulating Rho-kinase activation. Cell Commun Signal 2024; 22:58. [PMID: 38254202 PMCID: PMC10801962 DOI: 10.1186/s12964-023-01404-w] [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: 09/13/2023] [Accepted: 11/22/2023] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND The Ca2+-independent contraction of vascular smooth muscle is a leading cause of cardiovascular and cerebrovascular spasms. In the previous study, we demonstrated the involvement of Src family protein tyrosine kinase Fyn and Rho-kinase in the sphingosylphosphorylcholine (SPC)-induced abnormal and Ca2+-independent contraction of vascular smooth muscle, but the specific mechanism has not been completely clarified. METHODS Paxillin knockdown human coronary artery smooth muscle cells (CASMCs) and smooth muscle-specific paxillin knockout mice were generated by using paxillin shRNA and the tamoxifen-inducible Cre-LoxP system, respectively. CASMCs contraction was observed by time-lapse recording. The vessel contractility was measured by using a myography assay. Fyn, Rho-kinase, and myosin light chain activation were assessed by immunoprecipitation and western blotting. The paxillin expression and actin stress fibers were visualized by histological analysis and immunofluorescent staining. RESULTS The SPC-induced abnormal contraction was inhibited in paxillin knockdown CASMCs and arteries of paxillin knockout mice, indicating that paxillin is involved in this abnormal contraction. Further study showed that paxillin knockdown inhibited the SPC-induced Rho-kinase activation without affecting Fyn activation. In addition, paxillin knockdown significantly inhibited the SPC-induced actin stress fiber formation and myosin light chain phosphorylation. These results suggest that paxillin, as an upstream molecule of Rho-kinase, is involved in the SPC-induced abnormal contraction of vascular smooth muscle. CONCLUSIONS The present study demonstrated that paxillin participates in the SPC-induced abnormal vascular smooth muscle contraction by regulating Rho-kinase activation. Video Abstract.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Yamaguchi University, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan.
| | - Nan Li
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Yamaguchi University, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Sei Kobayashi
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Yamaguchi University, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan.
| |
Collapse
|
50
|
Liu H, Zhao Y, Zhao G, Deng Y, Chen YE, Zhang J. SWI/SNF Complex in Vascular Smooth Muscle Cells and Its Implications in Cardiovascular Pathologies. Cells 2024; 13:168. [PMID: 38247859 PMCID: PMC10814623 DOI: 10.3390/cells13020168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/09/2024] [Accepted: 01/12/2024] [Indexed: 01/23/2024] Open
Abstract
Mature vascular smooth muscle cells (VSMC) exhibit a remarkable degree of plasticity, a characteristic that has intrigued cardiovascular researchers for decades. Recently, it has become increasingly evident that the chromatin remodeler SWItch/Sucrose Non-Fermentable (SWI/SNF) complex plays a pivotal role in orchestrating chromatin conformation, which is critical for gene regulation. In this review, we provide a summary of research related to the involvement of the SWI/SNF complexes in VSMC and cardiovascular diseases (CVD), integrating these discoveries into the current landscape of epigenetic and transcriptional regulation in VSMC. These novel discoveries shed light on our understanding of VSMC biology and pave the way for developing innovative therapeutic strategies in CVD treatment.
Collapse
Affiliation(s)
- Hongyu Liu
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, 2800 Plymouth Road, Ann Arbor, MI 48109, USA; (H.L.); (Y.Z.)
- Department of Molecular & Integrative Physiology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
- Department of Vascular Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Yang Zhao
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, 2800 Plymouth Road, Ann Arbor, MI 48109, USA; (H.L.); (Y.Z.)
| | - Guizhen Zhao
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, 2800 Plymouth Road, Ann Arbor, MI 48109, USA; (H.L.); (Y.Z.)
| | - Yongjie Deng
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, 2800 Plymouth Road, Ann Arbor, MI 48109, USA; (H.L.); (Y.Z.)
| | - Y. Eugene Chen
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, 2800 Plymouth Road, Ann Arbor, MI 48109, USA; (H.L.); (Y.Z.)
- Department of Cardiac Surgery, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Jifeng Zhang
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, 2800 Plymouth Road, Ann Arbor, MI 48109, USA; (H.L.); (Y.Z.)
| |
Collapse
|