1
|
Zahradka P, Perera D, Charney J, Taylor CG. Distinct Effects of Wheat and Black Bean Consumption on Postprandial Vascular Responses in People with Arterial Stiffness: A Pilot Randomized Cross-Over Study. Nutrients 2025; 17:1159. [PMID: 40218917 PMCID: PMC11990509 DOI: 10.3390/nu17071159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 03/24/2025] [Accepted: 03/24/2025] [Indexed: 04/14/2025] Open
Abstract
Background/Objective: Postprandial vascular responses impact vascular health. This study investigated whether eating pulses or whole grains can acutely relax blood vessels in people with arterial stiffness. Methods: A single-blinded, controlled randomized cross-over clinical trial was conducted to compare the effects of pulses (¾ cup black beans) versus whole grains (¾ cup whole wheat kernels, also known as wheat berries) versus white rice (¾ cup) on postprandial vascular responses in males and females with established arterial stiffness (n = 9, 3M/6F, 50-64 years old). Peripheral and central hemodynamic measurements were obtained non-invasively prior to and 2 h after food consumption and were compared by t-test within a food type. Results: Peripheral and central systolic blood pressure was increased (4%) after eating white rice but not after the consumption of wheat or beans. A marked decline in augmentation index at 75 bpm (arterial stiffness) from 26.1 ± 3.6% to 16.2 ± 2.0% was observed 2 h after eating whole wheat but not beans or white rice. All foods slightly decreased heart rate at 2 h but had limited effects on other parameters of circulatory or heart health. Conclusions: Eating whole wheat or beans acutely improved overall vascular and heart health when compared to white rice. The effects of wheat and beans were distinct, with whole wheat having a major positive effect on blood vessel stiffness. The findings suggest that regular inclusion of both whole wheat and beans in the diet would be beneficial for improving cardiovascular health in persons exhibiting signs of arterial dysfunction, thus providing a potential therapeutic benefit for individuals who are at risk of heart attack and stroke. The study was registered (NCT05818358) on ClinicalTrials.gov.
Collapse
Affiliation(s)
- Peter Zahradka
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada; (P.Z.); (D.P.); (J.C.)
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Danielle Perera
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada; (P.Z.); (D.P.); (J.C.)
| | - Jordan Charney
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada; (P.Z.); (D.P.); (J.C.)
- Department of Foods and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Carla G. Taylor
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada; (P.Z.); (D.P.); (J.C.)
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- Department of Foods and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| |
Collapse
|
2
|
Ramirez-Perez FI, Jurrissen TJ, Augenreich MA, Castorena-Gonzalez JA, Morales-Quinones M, Foote CA, Nourian Z, Lateef OM, Imkaew N, Sun Z, Hill MA, Meininger GA, Padilla J, Martinez-Lemus LA. Small Arteries From Old Spontaneously Hypertensive Rats Exhibit Enhanced Endothelium-Independent Vasodilatory Capacity and Reduced Stiffness. Microcirculation 2025; 32:e70004. [PMID: 39962557 DOI: 10.1111/micc.70004] [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: 08/23/2024] [Revised: 12/23/2024] [Accepted: 02/02/2025] [Indexed: 05/09/2025]
Abstract
OBJECTIVE In conduit arteries, aging and hypertension are associated with stiffening characterized by increased cytoskeletal F-actin and endothelial dysfunction. Herein, we determined if this also happens at the level of the resistance vasculature. METHODS We retrospectively compared the mechanical and structural characteristics of small arteries isolated from older hypertensive and younger normotensive (64.7 ± 2.8 vs. 32.1 ± 1.9 years old) human subjects. The intersection of aging and hypertension was studied in small mesenteric arteries from old (88 weeks of age) spontaneously hypertensive (SHR) and Wistar Kyoto (WKY) normotensive rats. RESULTS Arteries from older hypertensive subjects were stiffer and had more F-actin, relative to those from younger normotensives. Comparatively, arteries from old SHRs showed reduced stiffness and increased vasodilation to sodium nitroprusside without changes in F-actin. Matrix metalloproteinase-2 (MMP-2) and -9 (MMP-9) were increased in the SHR arteries and exposure of naive arteries to exogenous MMP-2 and MMP-9 augmented responsiveness to sodium nitroprusside and adenosine. CONCLUSIONS In conclusion, resistance arteries from old SHRs are softer and vasodilate more to exogenous nitric oxide than those of WKY rats. This improved endothelial-independent vasodilation is associated with an increased vascular expression of MMP-2 and MMP-9. We further conclude that aging and hypertension effects on the microcirculation may vary between species and vascular beds.
Collapse
Affiliation(s)
| | - Thomas J Jurrissen
- NextGen Precision Health, University of Missouri, Columbia, Missouri, USA
| | - Marc A Augenreich
- NextGen Precision Health, University of Missouri, Columbia, Missouri, USA
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, USA
| | | | | | - Christopher A Foote
- NextGen Precision Health, University of Missouri, Columbia, Missouri, USA
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, USA
| | - Zahra Nourian
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, USA
| | - Olubodun M Lateef
- NextGen Precision Health, University of Missouri, Columbia, Missouri, USA
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, USA
| | - Natnicha Imkaew
- NextGen Precision Health, University of Missouri, Columbia, Missouri, USA
| | - Zhe Sun
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | - Michael A Hill
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | - Gerald A Meininger
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | - Jaume Padilla
- NextGen Precision Health, University of Missouri, Columbia, Missouri, USA
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
- Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri, USA
| | - Luis A Martinez-Lemus
- NextGen Precision Health, University of Missouri, Columbia, Missouri, USA
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
- Center for Precision Medicine, Department of Medicine, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
3
|
Arenas GA, Lorca RA. Effects of hypoxia on uteroplacental and fetoplacental vascular function during pregnancy. Front Physiol 2024; 15:1490154. [PMID: 39744703 PMCID: PMC11688409 DOI: 10.3389/fphys.2024.1490154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 12/03/2024] [Indexed: 02/13/2025] Open
Abstract
During pregnancy, marked changes in vasculature occur. The placenta is developed, and uteroplacental and fetoplacental circulations are established. These processes may be negatively affected by genetic anomalies, maternal environment (i.e., obesity or diabetes), and environmental conditions such as pollutants and hypoxia. Chronic hypoxia has detrimental effects on the vascular adaptations to pregnancy and fetal growth. The typical pregnancy-dependent rise in uterine blood flow by vascular remodeling and vasodilation of maternal uterine arteries is reduced, leading to increases in vascular tone. These maladaptations may lead to complications such as fetal growth restriction (FGR) and preeclampsia. In this review, the effect of hypoxia on uteroplacental and fetoplacental circulation and its impact on pregnancy outcomes in humans and animal models are discussed. Evidence is provided for several mechanisms that affect pregnancy through hypoxia-induced alterations. Future directions to fill gaps in knowledge and develop therapeutic strategies to prevent or alleviate hypoxia-related pregnancy complications, such as FGR and preeclampsia, are suggested.
Collapse
Affiliation(s)
| | - Ramón A. Lorca
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
4
|
Lateef OM, Foote C, Power G, Manrique-Acevedo C, Padilla J, Martinez-Lemus LA. LIM kinases in cardiovascular health and disease. Front Physiol 2024; 15:1506356. [PMID: 39744707 PMCID: PMC11688343 DOI: 10.3389/fphys.2024.1506356] [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: 10/05/2024] [Accepted: 11/28/2024] [Indexed: 01/14/2025] Open
Abstract
The Lim Kinase (LIMK) family of serine/threonine kinases is comprised of LIMK1 and LIMK2, which are central regulators of cytoskeletal dynamics via their well-characterized roles in promoting actin polymerization and destabilizing the cellular microtubular network. The LIMKs have been demonstrated to modulate several fundamental physiological processes, including cell cycle progression, cell motility and migration, and cell differentiation. These processes play important roles in maintaining cardiovascular health. However, LIMK activity in healthy and pathological states of the cardiovascular system is poorly characterized. This review highlights the cellular and molecular mechanisms involved in LIMK activation and inactivation, examining its roles in the pathophysiology of vascular and cardiac diseases such as hypertension, aneurysm, atrial fibrillation, and valvular heart disease. It addresses the LIMKs' involvement in processes that support cardiovascular health, including vasculogenesis, angiogenesis, and endothelial mechanotransduction. The review also features how LIMK activity participates in endothelial cell, vascular smooth muscle cell, and cardiomyocyte physiology and its implications in pathological states. A few recent preclinical studies demonstrate the therapeutic potential of LIMK inhibition. We conclude by proposing that future research should focus on the potential clinical relevance of LIMK inhibitors as therapeutic agents to reduce the burden of cardiovascular disease and improve patient outcomes.
Collapse
Affiliation(s)
- Olubodun M. Lateef
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
- Department of Medical Pharmacology and Physiology, University of Missouri Columbia, Columbia, MO, United States
| | - Christopher Foote
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
| | - Gavin Power
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Camila Manrique-Acevedo
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO, United States
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Columbia, MO, United States
- Center for Precision Medicine, Department of Medicine, University of Missouri, Columbia, MO, United States
| | - Jaume Padilla
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO, United States
| | - Luis A. Martinez-Lemus
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
- Department of Medical Pharmacology and Physiology, University of Missouri Columbia, Columbia, MO, United States
- Center for Precision Medicine, Department of Medicine, University of Missouri, Columbia, MO, United States
| |
Collapse
|
5
|
Wang Y, Chatterjee E, Li G, Xu J, Xiao J. Force-sensing protein expression in response to cardiovascular mechanotransduction. EBioMedicine 2024; 110:105412. [PMID: 39481337 PMCID: PMC11554632 DOI: 10.1016/j.ebiom.2024.105412] [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: 07/12/2024] [Revised: 10/01/2024] [Accepted: 10/07/2024] [Indexed: 11/02/2024] Open
Abstract
Force-sensing biophysical cues in microenvironment, including extracellular matrix performances, stretch-mediated mechanics, shear stress and flow-induced hemodynamics, have a significant influence in regulating vascular morphogenesis and cardiac remodeling by mechanotransduction. Once cells perceive these extracellular mechanical stimuli, Piezo activation promotes calcium influx by forming integrin-adhesion-coupling receptors. This induces robust contractility of cytoskeleton structures to further transmit biomechanical alternations into nuclei by regulating Hippo-Yes associated protein (YAP) signaling pathway between cytoplasmic and nuclear translocation. Although biomechanical stimuli are widely studied in cardiovascular diseases, the expression of force-sensing proteins in response to cardiovascular mechanotransduction has not been systematically concluded. Therefore, this review will summarize the force-sensing Piezo, cytoskeleton and YAP proteins to mediate extracellular mechanics, and also give the prominent emphasis on intrinsic connection of these mechanical proteins and cardiovascular mechanotransduction. Extensive insights into cardiovascular mechanics may provide some new strategies for cardiovascular clinical therapy.
Collapse
Affiliation(s)
- Yongtao Wang
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China; Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai University, Shanghai 200444, China
| | - Emeli Chatterjee
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Jiahong Xu
- Department of Cardiology, Shanghai Gongli Hospital, Shanghai 200135, China.
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China; Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
6
|
Ritsvall O, Albinsson S. Emerging role of YAP/TAZ in vascular mechanotransduction and disease. Microcirculation 2024; 31:e12838. [PMID: 38011540 DOI: 10.1111/micc.12838] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/08/2023] [Accepted: 11/13/2023] [Indexed: 11/29/2023]
Abstract
Cells have an incredible ability to physically interact with neighboring cells and their environment. They can detect and respond to mechanical forces by converting mechanical stimuli into biochemical signals in a process known as mechanotransduction. This is a key process for the adaption of vascular smooth muscle and endothelial cells to altered flow and pressure conditions. Mechanical stimuli, referring to a physical force exerted on cells, are primarily sensed by transmembrane proteins and the actin cytoskeleton, which initiate a cascade of intracellular events, including the activation of signaling pathways, ion channels, and transcriptional regulators. Recent work has highlighted an important role of the transcriptional coactivators YAP/TAZ for mechanotransduction in vascular cells. Interestingly, the activity of YAP/TAZ decreases with age, providing a potential mechanism for the detrimental effects of aging in the vascular wall. In this review, we summarize the current knowledge on the functional role of YAP and TAZ in vascular endothelial and smooth muscle cells for mechanotransduction in homeostasis and disease. In particular, the review is focused on in vivo observations from conditional knockout (KO) models of YAP/TAZ and the potential implications these studies may have for our understanding of vascular disease development.
Collapse
Affiliation(s)
- Olivia Ritsvall
- Department of Experimental Medical Science, Molecular Vascular Physiology, Lund University, Lund, Sweden
| | - Sebastian Albinsson
- Department of Experimental Medical Science, Molecular Vascular Physiology, Lund University, Lund, Sweden
| |
Collapse
|
7
|
Ferreira-Santos L, Martinez-Lemus LA, Padilla J. Sitting leg vasculopathy: potential adaptations beyond the endothelium. Am J Physiol Heart Circ Physiol 2024; 326:H760-H771. [PMID: 38241008 PMCID: PMC11221807 DOI: 10.1152/ajpheart.00489.2023] [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: 08/07/2023] [Revised: 12/27/2023] [Accepted: 01/18/2024] [Indexed: 02/29/2024]
Abstract
Increased sitting time, the most common form of sedentary behavior, is an independent risk factor for all-cause and cardiovascular disease mortality; however, the mechanisms linking sitting to cardiovascular risk remain largely elusive. Studies over the last decade have led to the concept that excessive time spent in the sitting position and the ensuing reduction in leg blood flow-induced shear stress cause endothelial dysfunction. This conclusion has been mainly supported by studies using flow-mediated dilation in the lower extremities as the measured outcome. In this review, we summarize evidence from classic studies and more recent ones that collectively support the notion that prolonged sitting-induced leg vascular dysfunction is likely also attributable to changes occurring in vascular smooth muscle cells (VSMCs). Indeed, we provide evidence that prolonged constriction of resistance arteries can lead to modifications in the structural characteristics of the vascular wall, including polymerization of actin filaments in VSMCs and inward remodeling, and that these changes manifest in a time frame that is consistent with the vascular changes observed with prolonged sitting. We expect this review will stimulate future studies with a focus on VSMC cytoskeletal remodeling as a potential target to prevent the detrimental vascular ramifications of too much sitting.
Collapse
Affiliation(s)
| | - Luis A Martinez-Lemus
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, United States
- Center for Precision Medicine, Department of Medicine, University of Missouri, Columbia, Missouri, United States
| | - Jaume Padilla
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States
- Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri, United States
| |
Collapse
|
8
|
Wang R, Huang R, Liu Y, Tamalunas A, Stief CG, Hennenberg M. Silencing of CDC42 inhibits contraction and growth-related functions in prostate stromal cells, which is mimicked by ML141. Life Sci 2023; 329:121928. [PMID: 37437651 DOI: 10.1016/j.lfs.2023.121928] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/06/2023] [Accepted: 07/08/2023] [Indexed: 07/14/2023]
Abstract
BACKGROUND Prostate smooth muscle contraction and stromal growth may contribute to lower urinary tract symptoms suggestive of benign prostatic hyperplasia, but are incompletely understood. A role of the monomeric GTPase CDC42 for smooth muscle contraction and proliferation appears possible, but is unknown for the prostate. Here, we silenced CDC42 expression in prostate stromal cells (WPMY-1), and examined contractility, growth-related functions and responses to the presumed CDC42 inhibitor, ML141. METHODS WPMY-1 cells were transfected with scrambled or CDC42-specific siRNA, and characterized for GTPase activities, contraction, proliferation, colony formation, apoptosis, cell death and viability. Effects of ML141 were examined in cells with and without silencing. RESULTS CDC42 silencing was confirmed by reduced mRNA and protein expression, and reduced CDC42 activity. Silencing impaired contraction (23-47 %), actin organization (25 %), proliferation (17-63 %), colony formation and viability (64-89 %), and increased the percentage of dead cells (2.6-fold). ML141 mimicked the phenotype of silencing in scrambled siRNA-transfected controls, and in non-transfected WPMY-1 cells, including inhibition of contraction, proliferation, colony formation and viability, breakdown of actin organization and increased cell death. In CDC42-silenced cells, ML141 still affected phalloiding organization, proliferation and cell death, with effect sizes resembling controls without silencing. ML141 inhibited RhoA activity in CDC42-silenced cells, but not in cells without silencing. CONCLUSIONS CDC42 promotes contraction of prostate stromal cells, and drives stromal growth by CDC42-mediated proliferation and suppression of apoptosis-independent cell death. ML141 mimicks all effects of CDC42 silencing, but its specificity may be limited and depends on GTPase phenotypes of cells.
Collapse
Affiliation(s)
- Ruixiao Wang
- Department of Urology, University Hospital Munich, LMU Munich, Munich, Germany
| | - Ru Huang
- Department of Urology, University Hospital Munich, LMU Munich, Munich, Germany
| | - Yuhan Liu
- Department of Urology, University Hospital Munich, LMU Munich, Munich, Germany
| | - Alexander Tamalunas
- Department of Urology, University Hospital Munich, LMU Munich, Munich, Germany
| | - Christan G Stief
- Department of Urology, University Hospital Munich, LMU Munich, Munich, Germany
| | - Martin Hennenberg
- Department of Urology, University Hospital Munich, LMU Munich, Munich, Germany.
| |
Collapse
|
9
|
Davis MJ, Earley S, Li YS, Chien S. Vascular mechanotransduction. Physiol Rev 2023; 103:1247-1421. [PMID: 36603156 PMCID: PMC9942936 DOI: 10.1152/physrev.00053.2021] [Citation(s) in RCA: 90] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 09/26/2022] [Accepted: 10/04/2022] [Indexed: 01/07/2023] Open
Abstract
This review aims to survey the current state of mechanotransduction in vascular smooth muscle cells (VSMCs) and endothelial cells (ECs), including their sensing of mechanical stimuli and transduction of mechanical signals that result in the acute functional modulation and longer-term transcriptomic and epigenetic regulation of blood vessels. The mechanosensors discussed include ion channels, plasma membrane-associated structures and receptors, and junction proteins. The mechanosignaling pathways presented include the cytoskeleton, integrins, extracellular matrix, and intracellular signaling molecules. These are followed by discussions on mechanical regulation of transcriptome and epigenetics, relevance of mechanotransduction to health and disease, and interactions between VSMCs and ECs. Throughout this review, we offer suggestions for specific topics that require further understanding. In the closing section on conclusions and perspectives, we summarize what is known and point out the need to treat the vasculature as a system, including not only VSMCs and ECs but also the extracellular matrix and other types of cells such as resident macrophages and pericytes, so that we can fully understand the physiology and pathophysiology of the blood vessel as a whole, thus enhancing the comprehension, diagnosis, treatment, and prevention of vascular diseases.
Collapse
Affiliation(s)
- Michael J Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Scott Earley
- Department of Pharmacology, University of Nevada, Reno, Nevada
| | - Yi-Shuan Li
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
| | - Shu Chien
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
- Department of Medicine, University of California, San Diego, California
| |
Collapse
|
10
|
Zhu Y, Chu Y, Wang S, Tang J, Li H, Feng L, Yu F, Ma X. Vascular Smooth Muscle TRPV4 (Transient Receptor Potential Vanilloid Family Member 4) Channels Regulate Vasoconstriction and Blood Pressure in Obesity. Hypertension 2023; 80:757-770. [PMID: 36794584 DOI: 10.1161/hypertensionaha.122.20109] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
BACKGROUND Vascular endothelium and smooth muscle work together to keep the balance of vasomotor tone and jointly maintain vascular homeostasis. Ca2+-permeable ion channel TRPV4 (transient receptor potential vanilloid family member 4) in endothelial cells regulates endothelium-dependent vasodilation and contraction in various states. However, how vascular smooth muscle cell TRPV4 (TRPV4SMC) contributes to vascular function and blood pressure regulation in physiological and pathologically obese condition has not been fully studied. METHODS We generated smooth muscle TRPV4-deficient mice and developed diet-induced obese mice model and analyzed the role of TRPV4SMC in intracellular Ca2+ ([Ca2+]i) regulation and vasoconstriction. Vasomotor changes of mouse mesenteric artery were measured by wire, and pressure myography. [Ca2+]i were measured by fluo-4 staining. Blood pressure was recorded by telemetric device. RESULTS Vascular TRPV4SMC played different roles in regulating vasomotor tone than endothelial TRPV4 due to their different features of [Ca2+]i regulation. Loss of TRPV4SMC attenuated U46619- and phenylephrine-induced contraction, suggesting its involvement in regulating vascular contractility. Mesenteric arteries from obese mice showed SMC hyperplasia, suggesting an increased level of TRPV4SMC. Loss of TRPV4SMC did not influence the development of obesity but protected mice from obesity-induced vasoconstriction and hypertension. In arteries deficient in SMC TRPV4, SMCs F-actin polymerization and RhoA dephosphorylation were attenuated under contractile stimuli. Moreover, SMC-dependent vasoconstriction was inhibited in human resistance arteries with TRPV4 inhibitor application. CONCLUSIONS Our data identify TRPV4SMC as a regulator of vascular contraction in both physiological states and pathologically obese mice. TRPV4SMC contributes to the ontogeny of vasoconstriction and hypertension induced by TRPV4SMC over-expression in obese mice mesenteric artery.
Collapse
Affiliation(s)
- Yifei Zhu
- Wuxi School of Medicine, Jiangnan University, Wuxi, China (Y.Z., Y.C., S.W., L.F., F.Y., X.M.)
| | - Yuan Chu
- Wuxi School of Medicine, Jiangnan University, Wuxi, China (Y.Z., Y.C., S.W., L.F., F.Y., X.M.)
| | - Sheng Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China (Y.Z., Y.C., S.W., L.F., F.Y., X.M.)
| | - Junjian Tang
- Department of Vascular Surgery, Affiliated Hospital of Jiangnan University, Wuxi, China (J.T., H.L.)
| | - Hu Li
- Department of Vascular Surgery, Affiliated Hospital of Jiangnan University, Wuxi, China (J.T., H.L.)
| | - Lei Feng
- Wuxi School of Medicine, Jiangnan University, Wuxi, China (Y.Z., Y.C., S.W., L.F., F.Y., X.M.)
| | - Fan Yu
- Wuxi School of Medicine, Jiangnan University, Wuxi, China (Y.Z., Y.C., S.W., L.F., F.Y., X.M.)
| | - Xin Ma
- Wuxi School of Medicine, Jiangnan University, Wuxi, China (Y.Z., Y.C., S.W., L.F., F.Y., X.M.)
| |
Collapse
|
11
|
Pharmacological targeting of the mitochondrial calcium-dependent potassium channel KCa3.1 triggers cell death and reduces tumor growth and metastasis in vivo. Cell Death Dis 2022; 13:1055. [PMID: 36539400 PMCID: PMC9768205 DOI: 10.1038/s41419-022-05463-8] [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: 07/27/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 12/24/2022]
Abstract
Ion channels are non-conventional, druggable oncological targets. The intermediate-conductance calcium-dependent potassium channel (KCa3.1) is highly expressed in the plasma membrane and in the inner mitochondrial membrane (mitoKCa3.1) of various cancer cell lines. The role mitoKCa3.1 plays in cancer cells is still undefined. Here we report the synthesis and characterization of two mitochondria-targeted novel derivatives of a high-affinity KCa3.1 antagonist, TRAM-34, which retain the ability to block channel activity. The effects of these drugs were tested in melanoma, pancreatic ductal adenocarcinoma and breast cancer lines, as well as in vivo in two orthotopic models. We show that the mitochondria-targeted TRAM-34 derivatives induce release of mitochondrial reactive oxygen species, rapid depolarization of the mitochondrial membrane, fragmentation of the mitochondrial network. They trigger cancer cell death with an EC50 in the µM range, depending on channel expression. In contrast, inhibition of the plasma membrane KCa3.1 by membrane-impermeant Maurotoxin is without effect, indicating a specific role of mitoKCa3.1 in determining cell fate. At sub-lethal concentrations, pharmacological targeting of mitoKCa3.1 significantly reduced cancer cell migration by enhancing production of mitochondrial reactive oxygen species and nuclear factor-κB (NF-κB) activation, and by downregulating expression of Bcl-2 Nineteen kD-Interacting Protein (BNIP-3) and of Rho GTPase CDC-42. This signaling cascade finally leads to cytoskeletal reorganization and impaired migration. Overexpression of BNIP-3 or pharmacological modulation of NF-κB and CDC-42 prevented the migration-reducing effect of mitoTRAM-34. In orthotopic models of melanoma and pancreatic ductal adenocarcinoma, the tumors at sacrifice were 60% smaller in treated versus untreated animals. Metastasis of melanoma cells to lymph nodes was also drastically reduced. No signs of toxicity were observed. In summary, our results identify mitochondrial KCa3.1 as an unexpected player in cancer cell migration and show that its pharmacological targeting is efficient against both tumor growth and metastatic spread in vivo.
Collapse
|
12
|
Deng Z, Zhang Y, Zhang Q, Li X, Zeng W, Jun C, Yuan D. Function of connexin 43 and RhoA/LIMK2/Cofilin signaling pathway in transient changes of contraction and dilation of human umbilical arterial smooth muscle cells. Int J Biochem Cell Biol 2022; 153:106326. [DOI: 10.1016/j.biocel.2022.106326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 10/21/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022]
|
13
|
Jurrissen TJ, Ramirez-Perez FI, Cabral-Amador FJ, Soares RN, Pettit-Mee RJ, Betancourt-Cortes EE, McMillan NJ, Sharma N, Rocha HNM, Fujie S, Morales-Quinones M, Lazo-Fernandez Y, Butler AA, Banerjee S, Sacks HS, Ibdah JA, Parks EJ, Rector RS, Manrique-Acevedo C, Martinez-Lemus LA, Padilla J. Role of adropin in arterial stiffening associated with obesity and type 2 diabetes. Am J Physiol Heart Circ Physiol 2022; 323:H879-H891. [PMID: 36083795 PMCID: PMC9602697 DOI: 10.1152/ajpheart.00385.2022] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/06/2022] [Accepted: 09/06/2022] [Indexed: 01/16/2023]
Abstract
Adropin is a peptide largely secreted by the liver and known to regulate energy homeostasis; however, it also exerts cardiovascular effects. Herein, we tested the hypothesis that low circulating levels of adropin in obesity and type 2 diabetes (T2D) contribute to arterial stiffening. In support of this hypothesis, we report that obesity and T2D are associated with reduced levels of adropin (in liver and plasma) and increased arterial stiffness in mice and humans. Establishing causation, we show that mesenteric arteries from adropin knockout mice are also stiffer, relative to arteries from wild-type counterparts, thus recapitulating the stiffening phenotype observed in T2D db/db mice. Given the above, we performed a set of follow-up experiments, in which we found that 1) exposure of endothelial cells or isolated mesenteric arteries from db/db mice to adropin reduces filamentous actin (F-actin) stress fibers and stiffness, 2) adropin-induced reduction of F-actin and stiffness in endothelial cells and db/db mesenteric arteries is abrogated by inhibition of nitric oxide (NO) synthase, and 3) stimulation of smooth muscle cells or db/db mesenteric arteries with a NO mimetic reduces stiffness. Lastly, we demonstrated that in vivo treatment of db/db mice with adropin for 4 wk reduces stiffness in mesenteric arteries. Collectively, these findings indicate that adropin can regulate arterial stiffness, likely via endothelium-derived NO, and thus support the notion that "hypoadropinemia" should be considered as a putative target for the prevention and treatment of arterial stiffening in obesity and T2D.NEW & NOTEWORTHY Arterial stiffening, a characteristic feature of obesity and type 2 diabetes (T2D), contributes to the development and progression of cardiovascular diseases. Herein we establish that adropin is decreased in obese and T2D models and furthermore provide evidence that reduced adropin may directly contribute to arterial stiffening. Collectively, findings from this work support the notion that "hypoadropinemia" should be considered as a putative target for the prevention and treatment of arterial stiffening in obesity and T2D.
Collapse
Affiliation(s)
- Thomas J Jurrissen
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | | | | | - Rogerio N Soares
- Department of Medicine, Center for Precision Medicine, University of Missouri, Columbia, Missouri
| | - Ryan J Pettit-Mee
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | | | - Neil J McMillan
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - Neekun Sharma
- Department of Medicine, Center for Precision Medicine, University of Missouri, Columbia, Missouri
| | - Helena N M Rocha
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
- Department of Physiology and Pharmacology, Fluminense Federal University, Niteroi, Brazil
| | - Shumpei Fujie
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
- Faculty of Sport and Health Science, Ritsumeikan University, Shiga, Japan
| | - Mariana Morales-Quinones
- Department of Medicine, Center for Precision Medicine, University of Missouri, Columbia, Missouri
| | - Yoskaly Lazo-Fernandez
- Department of Medicine, Center for Precision Medicine, University of Missouri, Columbia, Missouri
| | - Andrew A Butler
- Department of Pharmacology and Physiological Sciences, Saint Louis University, Saint Louis, Missouri
| | - Subhashis Banerjee
- Department of Pharmacology and Physiological Sciences, Saint Louis University, Saint Louis, Missouri
| | - Harold S Sacks
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Jamal A Ibdah
- Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, Missouri
| | - Elizabeth J Parks
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, Missouri
| | - R Scott Rector
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
- Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, Missouri
| | - Camila Manrique-Acevedo
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
- Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, Missouri
| | - Luis A Martinez-Lemus
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
- Department of Medicine, Center for Precision Medicine, University of Missouri, Columbia, Missouri
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Jaume Padilla
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
- Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri
| |
Collapse
|
14
|
Daoud F, Arévalo Martinez M, Holmberg J, Alajbegovic A, Ali N, Rippe C, Swärd K, Albinsson S. YAP and TAZ in Vascular Smooth Muscle Confer Protection Against Hypertensive Vasculopathy. Arterioscler Thromb Vasc Biol 2022; 42:428-443. [PMID: 35196875 PMCID: PMC8939708 DOI: 10.1161/atvbaha.121.317365] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Hypertension remains a major risk factor for cardiovascular diseases, but the underlying mechanisms are not well understood. We hypothesize that appropriate mechanotransduction and contractile function in vascular smooth muscle cells are crucial to maintain vascular wall integrity. The Hippo pathway effectors YAP (yes-associated protein 1) and TAZ (WW domain containing transcription regulator 1) have been identified as mechanosensitive transcriptional coactivators. However, their role in vascular smooth muscle cell mechanotransduction has not been investigated in vivo. METHODS We performed physiological and molecular analyses utilizing an inducible smooth muscle-specific YAP/TAZ knockout mouse model. RESULTS Arteries lacking YAP/TAZ have reduced agonist-mediated contraction, decreased myogenic response, and attenuated stretch-induced transcriptional regulation of smooth muscle markers. Moreover, in established hypertension, YAP/TAZ knockout results in severe vascular lesions in small mesenteric arteries characterized by neointimal hyperplasia, elastin degradation, and adventitial thickening. CONCLUSIONS This study demonstrates a protective role of YAP/TAZ against hypertensive vasculopathy.
Collapse
Affiliation(s)
- Fatima Daoud
- Department of Experimental Medical Science (F.D., M.A.M., J.H., A.A., C.R., K.S., S.A.), Lund University, Sweden
| | - Marycarmen Arévalo Martinez
- Department of Experimental Medical Science (F.D., M.A.M., J.H., A.A., C.R., K.S., S.A.), Lund University, Sweden
| | - Johan Holmberg
- Department of Experimental Medical Science (F.D., M.A.M., J.H., A.A., C.R., K.S., S.A.), Lund University, Sweden
| | - Azra Alajbegovic
- Department of Experimental Medical Science (F.D., M.A.M., J.H., A.A., C.R., K.S., S.A.), Lund University, Sweden
| | - Neserin Ali
- Department of Clinical Sciences Lund, Orthopaedics, Clinical Epidemiology Unit (N.A.), Lund University, Sweden
| | - Catarina Rippe
- Department of Experimental Medical Science (F.D., M.A.M., J.H., A.A., C.R., K.S., S.A.), Lund University, Sweden
| | - Karl Swärd
- Department of Experimental Medical Science (F.D., M.A.M., J.H., A.A., C.R., K.S., S.A.), Lund University, Sweden
| | - Sebastian Albinsson
- Department of Experimental Medical Science (F.D., M.A.M., J.H., A.A., C.R., K.S., S.A.), Lund University, Sweden
| |
Collapse
|
15
|
Horn AG, Kunkel ON, Baumfalk DR, Simon ME, Schulze KM, Hsu WW, Muller-Delp J, Poole DC, Behnke BJ. Prolonged mechanical ventilation increases diaphragm arteriole circumferential stretch without changes in stress/stretch: Implications for the pathogenesis of ventilator-induced diaphragm dysfunction. Microcirculation 2021; 28:e12727. [PMID: 34467606 DOI: 10.1111/micc.12727] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 05/31/2021] [Accepted: 08/24/2021] [Indexed: 01/27/2023]
Abstract
INTRODUCTION Prolonged mechanical ventilation (MV; ≥6 h) results in large, time-dependent reductions in diaphragmatic blood flow and shear stress. We tested the hypothesis that MV would impair the structural and material properties (ie, increased stress/stretch relation and/or circumferential stretch) of first-order arterioles (1A) from the medial costal diaphragm. METHODS Shear stress was estimated from isolated arterioles and prior blood flow data from the diaphragm during spontaneous breathing (SB) and prolonged MV (6 h MV). Thereafter, female Sprague-Dawley rats (~5 months) were randomly divided into two groups, SB (n = 6) and 6 h MV (n = 6). Following SB and 6 h MV, 1A medial costal diaphragm arterioles were isolated, cannulated, and subjected to stepwise (0-140 cmH2 O) increases in intraluminal pressure in calcium-free Ringer's solution. Inner diameter and wall thickness were measured at each pressure step and used to calculate wall:lumen ratio, Cauchy-stress, and circumferential stretch. RESULTS Compared to SB, there was a ~90% reduction in arteriolar shear stress with prolonged MV (9 ± 2 vs 78 ± 20 dynes/cm2 ; p ≤ .05). In the unloaded condition (0 cmH2 O), the arteriolar intraluminal diameter was reduced (37 ± 8 vs 79 ± 13 μm) and wall:lumen ratio was increased (120 ± 18 vs 46 ± 10%) compared to SB (p ≤ .05). There were no differences in the passive diameter responses or the circumferential stress/stretch relationship between groups (p > .05), but at each pressure step, circumferential stretch was increased with 6 h MV vs SB (p ≤ .05). CONCLUSION During prolonged MV, medial costal diaphragm arteriolar shear stress is severely diminished. Despite no change in the material behavior (stress/stretch), prolonged MV resulted in altered structural and mechanical properties (ie, elevated circumferential stretch) of medial costal diaphragm arterioles. This provides important novel mechanistic insights into the impaired diaphragm blood flow capacity and vascular dysfunction following prolonged MV.
Collapse
Affiliation(s)
- Andrew G Horn
- Department of Kinesiology, Kansas State University, Manhattan, KS, USA
| | - Olivia N Kunkel
- Department of Kinesiology, Kansas State University, Manhattan, KS, USA
| | - Dryden R Baumfalk
- Department of Kinesiology, Kansas State University, Manhattan, KS, USA
| | - Mikaela E Simon
- Department of Kinesiology, Kansas State University, Manhattan, KS, USA
| | - Kiana M Schulze
- Department of Kinesiology, Kansas State University, Manhattan, KS, USA
| | - Wei-Wen Hsu
- Division of Biostatistics and Bioinformations, Department of Environmental and Public Health Sciences, University of Cincinnati, Cincinnati, OH, USA
| | - Judy Muller-Delp
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL, USA
| | - David C Poole
- Department of Kinesiology, Kansas State University, Manhattan, KS, USA
- Department of Anatomy and Physiology, Kansas State University, Manhattan, KS, USA
| | - Bradley J Behnke
- Department of Kinesiology, Kansas State University, Manhattan, KS, USA
- Johnson Cancer Research Center, Kansas State University, Manhattan, KS, USA
| |
Collapse
|
16
|
Wenceslau CF, McCarthy CG, Earley S, England SK, Filosa JA, Goulopoulou S, Gutterman DD, Isakson BE, Kanagy NL, Martinez-Lemus LA, Sonkusare SK, Thakore P, Trask AJ, Watts SW, Webb RC. Guidelines for the measurement of vascular function and structure in isolated arteries and veins. Am J Physiol Heart Circ Physiol 2021; 321:H77-H111. [PMID: 33989082 PMCID: PMC8321813 DOI: 10.1152/ajpheart.01021.2020] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 04/26/2021] [Accepted: 05/11/2021] [Indexed: 12/11/2022]
Abstract
The measurement of vascular function in isolated vessels has revealed important insights into the structural, functional, and biomechanical features of the normal and diseased cardiovascular system and has provided a molecular understanding of the cells that constitutes arteries and veins and their interaction. Further, this approach has allowed the discovery of vital pharmacological treatments for cardiovascular diseases. However, the expansion of the vascular physiology field has also brought new concerns over scientific rigor and reproducibility. Therefore, it is appropriate to set guidelines for the best practices of evaluating vascular function in isolated vessels. These guidelines are a comprehensive document detailing the best practices and pitfalls for the assessment of function in large and small arteries and veins. Herein, we bring together experts in the field of vascular physiology with the purpose of developing guidelines for evaluating ex vivo vascular function. By using this document, vascular physiologists will have consistency among methodological approaches, producing more reliable and reproducible results.
Collapse
Grants
- R01HL139585 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- P20 GM130459 NIGMS NIH HHS
- R01HL121871 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- DK115255 HHS | NIH | National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
- R61 NS115132 NINDS NIH HHS
- K99HL151889 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL151413 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R00HL116769 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL091905 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL088554 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL139585 NHLBI NIH HHS
- P20GM130459 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- R01 HL135901 NHLBI NIH HHS
- RF1 NS110044 NINDS NIH HHS
- R01ES014639 HHS | NIH | National Institute of Environmental Health Sciences (NIEHS)
- U24 DK076169 NIDDK NIH HHS
- S10OD023438 HHS | NIH | NIH Office of the Director (OD)
- R01HL137112 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL135901 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL146914 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R00 HL116769 NHLBI NIH HHS
- K99 HL151889 NHLBI NIH HHS
- U24 DK115255 NIDDK NIH HHS
- R21 EB026518 NIBIB NIH HHS
- R01 HL149762 NHLBI NIH HHS
- DK076169 HHS | NIH | National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
- R01NS082521 HHS | NIH | National Institute of Neurological Disorders and Stroke (NINDS)
- R01 HL146054 NHLBI NIH HHS
- R21EB026518 HHS | NIH | National Institute of Biomedical Imaging and Bioengineering (NIBIB)
- R01 HL123301 NHLBI NIH HHS
- P01 HL134604 NHLBI NIH HHS
- R00GM118885 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- R01 HL091905 NHLBI NIH HHS
- RF1NS110044 HHS | NIH | National Institute of Neurological Disorders and Stroke (NINDS)
- R01HL142808 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R61NS115132 HHS | NIH | National Institute of Neurological Disorders and Stroke (NINDS)
- R01HL146562 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL088105 NHLBI NIH HHS
- SB1 HL121871 NHLBI NIH HHS
- R01 HD037831 NICHD NIH HHS
- R01 HL137852 NHLBI NIH HHS
- R35 HL155008 NHLBI NIH HHS
- R01 HL137112 NHLBI NIH HHS
- R01HL149762 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL123301 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL146914 NHLBI NIH HHS
- R01 HL142808 NHLBI NIH HHS
- R01 HL088554 NHLBI NIH HHS
- R01HD037831 HHS | NIH | Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD)
- R01HL146054 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL146562 NHLBI NIH HHS
- R44 HL121871 NHLBI NIH HHS
- R01HL088105 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 ES014639 NIEHS NIH HHS
- P01HL134604 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL137852 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- S10 OD023438 NIH HHS
- R01 HL151413 NHLBI NIH HHS
- R41 HL121871 NHLBI NIH HHS
- R00 GM118885 NIGMS NIH HHS
Collapse
Affiliation(s)
- Camilla F Wenceslau
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Cameron G McCarthy
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Scott Earley
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, Reno School of Medicine, University of Nevada, Reno, Nevada
| | - Sarah K England
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri
| | - Jessica A Filosa
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Styliani Goulopoulou
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - David D Gutterman
- Department of Medicine, Medical College of Wisconsin Cardiovascular Center, Milwaukee, Wisconsin
| | - Brant E Isakson
- Department of Molecular Physiology and Biophysics, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Nancy L Kanagy
- Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, New Mexico
| | - Luis A Martinez-Lemus
- Department of Medical Pharmacology and Physiology, Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Swapnil K Sonkusare
- Department of Molecular Physiology and Biophysics, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Pratish Thakore
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, Reno School of Medicine, University of Nevada, Reno, Nevada
| | - Aaron J Trask
- Center for Cardiovascular Research, The Heart Center, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio
| | - Stephanie W Watts
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - R Clinton Webb
- Cardiovascular Translational Research Center, Department of Cell Biology and Anatomy, University of South Carolina, Columbia, South Carolina
| |
Collapse
|
17
|
Liao J, Lu W, Chen Y, Duan X, Zhang C, Luo X, Lin Z, Chen J, Liu S, Yan H, Chen Y, Feng H, Zhou D, Chen X, Zhang Z, Yang Q, Liu X, Tang H, Li J, Makino A, Yuan JXJ, Zhong N, Yang K, Wang J. Upregulation of Piezo1 (Piezo Type Mechanosensitive Ion Channel Component 1) Enhances the Intracellular Free Calcium in Pulmonary Arterial Smooth Muscle Cells From Idiopathic Pulmonary Arterial Hypertension Patients. Hypertension 2021; 77:1974-1989. [PMID: 33813851 DOI: 10.1161/hypertensionaha.120.16629] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Jing Liao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Wenju Lu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Yuqin Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Xin Duan
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (X.D.)
| | - Chenting Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Xiaoyun Luo
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Ziying Lin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Jiyuan Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.).,Department of Medicine, University of California, San Diego, La Jolla (J.C., A.M., J.X.-J.Y., J.W.)
| | - Shiyun Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Han Yan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Yilin Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Huazhuo Feng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Dansha Zhou
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Xu Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Zizhou Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Qifeng Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Xinyi Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Haiyang Tang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Jing Li
- Lingnan Medical Research Center, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, China (J. Li)
| | - Ayako Makino
- Department of Medicine, University of California, San Diego, La Jolla (J.C., A.M., J.X.-J.Y., J.W.)
| | - Jason X-J Yuan
- Department of Medicine, University of California, San Diego, La Jolla (J.C., A.M., J.X.-J.Y., J.W.)
| | - Nanshan Zhong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Kai Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Jian Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.).,Department of Medicine, University of California, San Diego, La Jolla (J.C., A.M., J.X.-J.Y., J.W.)
| |
Collapse
|
18
|
Clark JL, Taylor CG, Zahradka P. Black beans and red kidney beans induce positive postprandial vascular responses in healthy adults: A pilot randomized cross-over study. Nutr Metab Cardiovasc Dis 2021; 31:216-226. [PMID: 32917495 DOI: 10.1016/j.numecd.2020.07.042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/15/2020] [Accepted: 07/25/2020] [Indexed: 11/18/2022]
Abstract
BACKGROUND AND AIMS Consuming pulses (dry beans, dry peas, chickpeas, lentils) over several weeks can improve vascular function and decrease cardiovascular disease risk; however, it is unknown whether pulses can modulate postprandial vascular responses. The objective of this study was to compare different bean varieties (black, navy, pinto, red kidney) and white rice for their acute postprandial effects on vascular and metabolic responses in healthy individuals. METHODS AND RESULTS The study was designed as a single-blinded, randomized crossover trial with a minimum 6 days between consumption of the food articles. Vascular tone (primary endpoint), haemodynamics and serum biochemistry (secondary endpoints) were measured in 8 healthy adults before and at 1, 2, and 6 h after eating ¾ cup of beans or rice. Blood pressure and pulse wave velocity (PWV) were lower at 2 h following red kidney bean and pinto bean consumption compared to rice and navy bean, respectively (p < 0.05). There was greater vasorelaxation 6 h following consumption of darker-coloured beans, as shown by decreased vascular tone: PWV was lower after consuming black bean compared to pinto bean, augmentation pressure was lower after consuming black bean compared to rice and pinto bean, and wave reflection magnitude was lower after consuming red kidney bean and black bean compared to rice, navy bean, and pinto bean (p < 0.05). LDL-cholesterol concentrations were lower 6 h after black bean consumption compared to rice (p < 0.05). CONCLUSION Overall, red kidney and black beans, the darker-coloured beans, elicited a positive effect on the tensile properties of blood vessels, and this acute response may provide insight for how pulses modify vascular function.
Collapse
Affiliation(s)
- Jaime L Clark
- Department of Food and Human Nutritional Sciences, Faculty of Agricultural and Food Sciences, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada; Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, R2H 2A6, Canada.
| | - Carla G Taylor
- Department of Food and Human Nutritional Sciences, Faculty of Agricultural and Food Sciences, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada; Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, R2H 2A6, Canada; Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, R3E 0T5, Canada.
| | - Peter Zahradka
- Department of Food and Human Nutritional Sciences, Faculty of Agricultural and Food Sciences, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada; Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, R2H 2A6, Canada; Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, R3E 0T5, Canada.
| |
Collapse
|
19
|
Bajpai A, Li R, Chen W. The cellular mechanobiology of aging: from biology to mechanics. Ann N Y Acad Sci 2020; 1491:3-24. [PMID: 33231326 DOI: 10.1111/nyas.14529] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 10/10/2020] [Accepted: 10/21/2020] [Indexed: 12/14/2022]
Abstract
Aging is a chronic, complicated process that leads to degenerative physical and biological changes in living organisms. Aging is associated with permanent, gradual physiological cellular decay that affects all aspects of cellular mechanobiological features, including cellular cytoskeleton structures, mechanosensitive signaling pathways, and forces in the cell, as well as the cell's ability to sense and adapt to extracellular biomechanical signals in the tissue environment through mechanotransduction. These mechanobiological changes in cells are directly or indirectly responsible for dysfunctions and diseases in various organ systems, including the cardiovascular, musculoskeletal, skin, and immune systems. This review critically examines the role of aging in the progressive decline of the mechanobiology occurring in cells, and establishes mechanistic frameworks to understand the mechanobiological effects of aging on disease progression and to develop new strategies for halting and reversing the aging process. Our review also highlights the recent development of novel bioengineering approaches for studying the key mechanobiological mechanisms in aging.
Collapse
Affiliation(s)
- Apratim Bajpai
- Department of Mechanical and Aerospace Engineering, Tandon School of Engineering, New York University, Brooklyn, New York
| | - Rui Li
- Department of Mechanical and Aerospace Engineering, Tandon School of Engineering, New York University, Brooklyn, New York.,Department of Biomedical Engineering, Tandon School of Engineering, New York University, Brooklyn, New York
| | - Weiqiang Chen
- Department of Mechanical and Aerospace Engineering, Tandon School of Engineering, New York University, Brooklyn, New York.,Department of Biomedical Engineering, Tandon School of Engineering, New York University, Brooklyn, New York.,Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York
| |
Collapse
|
20
|
Takaishi K, Kinoshita H, Feng GG, Azma T, Kawahito S, Kitahata H. Cytoskeleton-disrupting agent cytochalasin B reduces oxidative stress caused by high glucose in the human arterial smooth muscle. J Pharmacol Sci 2020; 144:197-203. [PMID: 33070838 DOI: 10.1016/j.jphs.2020.08.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 08/01/2020] [Accepted: 08/19/2020] [Indexed: 12/15/2022] Open
Abstract
The role of cytoskeleton dynamics in the oxidative stress toward human vasculature has been unclear. The current study examined whether the cytoskeleton-disrupting agent cytochalasin B reduces oxidative stress caused by high glucose in the human arterial smooth muscle. All experiments in the human omental arteries without endothelium or the cultured human coronary artery smooth muscle cells were performed in d-glucose (5.5 mmol/L). The exposure toward d-glucose (20 mmol/L) for 60 min reduced the relaxation or hyperpolarization to an ATP sensitive K+ channel (KATP) opener levcromakalim (10-8 to 3 × 10-6 mol/L and 3 × 10-6 mol/L, respectively). Cytochalasin B and a superoxide inhibitor Tiron, restored them similarly. Cytochalasin B reduced the NADPH oxidase activity, leading to a decrease in superoxide levels of the arteries treated with high d-glucose. Also, cytochalasin B impaired the F-actin constitution and the membrane translocation of an NADPH oxidase subunit p47phox in artery smooth muscle cells treated with high d-glucose. A clinical concentration of cytochalasin B prevented human vascular smooth muscle malfunction via the oxidative stress caused by high glucose. Regulation of the cytoskeleton may be essential to keep the normal vascular function in patients with hyperglycemia.
Collapse
Affiliation(s)
- Kazumi Takaishi
- Department of Dental Anesthesiology, Tokushima University Hospital, Tokushima, Japan
| | - Hiroyuki Kinoshita
- Department of Anesthesiology, Tokushima University Hospital, Tokushima, Japan; Department of Anesthesiology, Aichi Medical University School of Medicine, Aichi, Japan.
| | - Guo-Gang Feng
- Department of Anesthesiology, Aichi Medical University School of Medicine, Aichi, Japan
| | - Toshiharu Azma
- Department of Anesthesiology & Pain Medicine, Kohnodai Hospital, National Center for Global Health and Medicine, Ichikawa, Japan
| | - Shinji Kawahito
- Department of Anesthesiology, Tokushima University Hospital, Tokushima, Japan
| | - Hiroshi Kitahata
- Department of Dental Anesthesiology, Tokushima University Hospital, Tokushima, Japan
| |
Collapse
|
21
|
Joe B, McCarthy CG, Edwards JM, Cheng X, Chakraborty S, Yang T, Golonka RM, Mell B, Yeo JY, Bearss NR, Furtado J, Saha P, Yeoh BS, Vijay-Kumar M, Wenceslau CF. Microbiota Introduced to Germ-Free Rats Restores Vascular Contractility and Blood Pressure. Hypertension 2020; 76:1847-1855. [PMID: 33070663 DOI: 10.1161/hypertensionaha.120.15939] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Commensal gut microbiota are strongly correlated with host hemodynamic homeostasis but only broadly associated with cardiovascular health. This includes a general correspondence of quantitative and qualitative shifts in intestinal microbial communities found in hypertensive rat models and human patients. However, the mechanisms by which gut microbes contribute to the function of organs important for blood pressure (BP) control remain unanswered. To examine the direct effects of microbiota on BP, we conventionalized germ-free (GF) rats with specific pathogen-free rats for a short-term period of 10 days, which served as a model system to observe the dynamic responses when reconstituting the holobiome. The absence of microbiota in GF rats resulted with relative hypotension compared with their conventionalized counterparts, suggesting an obligatory role of microbiota in BP homeostasis. Hypotension observed in GF rats was accompanied by a marked reduction in vascular contractility. Both BP and vascular contractility were restored by the introduction of microbiota to GF rats, indicating that microbiota could impact BP through a vascular-dependent mechanism. This is further supported by the decrease in actin polymerization in arteries from GF rats. Improved vascular contractility in conventionalized GF rats, as indicated through stabilized actin filaments, was associated with an increase in cofilin phosphorylation. These data indicate that the vascular system senses the presence (or lack of) microbiota to maintain vascular tone via actin polymerization. Overall, these results constitute a fundamental discovery of the essential nature of microbiota in BP regulation.
Collapse
Affiliation(s)
- Bina Joe
- From the UT Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Cameron G McCarthy
- From the UT Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Jonnelle M Edwards
- From the UT Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Xi Cheng
- From the UT Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Saroj Chakraborty
- From the UT Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Tao Yang
- From the UT Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Rachel M Golonka
- From the UT Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Blair Mell
- From the UT Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Ji-Youn Yeo
- From the UT Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Nicole R Bearss
- From the UT Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Janara Furtado
- From the UT Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Piu Saha
- From the UT Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Beng San Yeoh
- From the UT Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Matam Vijay-Kumar
- From the UT Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Camilla F Wenceslau
- From the UT Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| |
Collapse
|
22
|
Li B, Wang R, Wang Y, Stief CG, Hennenberg M. Regulation of smooth muscle contraction by monomeric non-RhoA GTPases. Br J Pharmacol 2020; 177:3865-3877. [PMID: 32579705 PMCID: PMC7429483 DOI: 10.1111/bph.15172] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/05/2020] [Accepted: 06/09/2020] [Indexed: 12/13/2022] Open
Abstract
Smooth muscle contraction in the cardiovascular system, airways, prostate and lower urinary tract is involved in the pathophysiology of many diseases, including cardiovascular and obstructive lung disease plus lower urinary tract symptoms, which are associated with high prevalence of morbidity and mortality. This prominent clinical role of smooth muscle tone has led to the molecular mechanisms involved being subjected to extensive research. In general smooth muscle contraction is promoted by three major signalling pathways, including the monomeric GTPase RhoA pathway. However, emerging evidence suggests that monomeric GTPases other than RhoA may be involved in signal transduction in smooth muscle contraction, including Rac GTPases, cell division control protein 42 homologue, adenosine ribosylation factor 6, Ras, Rap1b and Rab GTPases. Here, we review these emerging functions of non-RhoA GTPases in smooth muscle contraction, which has now become increasingly more evident and constitutes an emerging and innovative research area of high clinical relevance.
Collapse
Affiliation(s)
- Bingsheng Li
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | - Ruixiao Wang
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | - Yiming Wang
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | - Christian G Stief
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | - Martin Hennenberg
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
23
|
Morales-Quinones M, Ramirez-Perez FI, Foote CA, Ghiarone T, Ferreira-Santos L, Bloksgaard M, Spencer N, Kimchi ET, Manrique-Acevedo C, Padilla J, Martinez-Lemus LA. LIMK (LIM Kinase) Inhibition Prevents Vasoconstriction- and Hypertension-Induced Arterial Stiffening and Remodeling. Hypertension 2020; 76:393-403. [PMID: 32594801 DOI: 10.1161/hypertensionaha.120.15203] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Increased arterial stiffness and vascular remodeling precede and are consequences of hypertension. They also contribute to the development and progression of life-threatening cardiovascular diseases. Yet, there are currently no agents specifically aimed at preventing or treating arterial stiffening and remodeling. Previous research indicates that vascular smooth muscle actin polymerization participates in the initial stages of arterial stiffening and remodeling and that LIMK (LIM kinase) promotes F-actin formation and stabilization via cofilin phosphorylation and consequent inactivation. Herein, we hypothesize that LIMK inhibition is able to prevent vasoconstriction- and hypertension-associated arterial stiffening and inward remodeling. We found that small visceral arteries isolated from hypertensive subjects are stiffer and have greater cofilin phosphorylation than those from nonhypertensives. We also show that LIMK inhibition prevents arterial stiffening and inward remodeling in isolated human small visceral arteries exposed to prolonged vasoconstriction. Using cultured vascular smooth muscle cells, we determined that LIMK inhibition prevents vasoconstrictor agonists from increasing cofilin phosphorylation, F-actin volume, and cell cortex stiffness. We further show that localized LIMK inhibition prevents arteriolar inward remodeling in hypertensive mice. This indicates that hypertension is associated with increased vascular smooth muscle cofilin phosphorylation, cytoskeletal stress fiber formation, and heightened arterial stiffness. Our data further suggest that pharmacological inhibition of LIMK prevents vasoconstriction-induced arterial stiffening, in part, via reductions in vascular smooth muscle F-actin content and cellular stiffness. Accordingly, LIMK inhibition should represent a promising therapeutic means to stop the progression of arterial stiffening and remodeling in hypertension.
Collapse
Affiliation(s)
- Mariana Morales-Quinones
- From the Dalton Cardiovascular Research Center (M.M.-Q., F.I.R.-P., C.A.F., T.G., L.F.-S., C.M.-A., J.P., L.A.M.-L.), University of Missouri, Columbia, MO
| | - Francisco I Ramirez-Perez
- From the Dalton Cardiovascular Research Center (M.M.-Q., F.I.R.-P., C.A.F., T.G., L.F.-S., C.M.-A., J.P., L.A.M.-L.), University of Missouri, Columbia, MO.,Department of Biological Engineering (F.I.R.-P., L.A.M.-L.), University of Missouri, Columbia, MO
| | - Christopher A Foote
- From the Dalton Cardiovascular Research Center (M.M.-Q., F.I.R.-P., C.A.F., T.G., L.F.-S., C.M.-A., J.P., L.A.M.-L.), University of Missouri, Columbia, MO
| | - Thaysa Ghiarone
- From the Dalton Cardiovascular Research Center (M.M.-Q., F.I.R.-P., C.A.F., T.G., L.F.-S., C.M.-A., J.P., L.A.M.-L.), University of Missouri, Columbia, MO
| | - Larissa Ferreira-Santos
- From the Dalton Cardiovascular Research Center (M.M.-Q., F.I.R.-P., C.A.F., T.G., L.F.-S., C.M.-A., J.P., L.A.M.-L.), University of Missouri, Columbia, MO.,Instituto do Coração (InCor), Hospital das Clinicas, Faculdade de Medicina, Universidade de São Paulo, Brazil (L.F.-S.)
| | - Maria Bloksgaard
- Department of Molecular Medicine, University of Southern Denmark, Odense (M.B.)
| | | | - Eric T Kimchi
- Department of Surgery (E.T.K.), University of Missouri, Columbia, MO.,Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO (E.T.K., C.M.-A.)
| | - Camila Manrique-Acevedo
- From the Dalton Cardiovascular Research Center (M.M.-Q., F.I.R.-P., C.A.F., T.G., L.F.-S., C.M.-A., J.P., L.A.M.-L.), University of Missouri, Columbia, MO.,Department of Medicine, Division of Endocrinology, Diabetes and Metabolism (C.M.-A.), University of Missouri, Columbia, MO.,Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO (E.T.K., C.M.-A.)
| | - Jaume Padilla
- From the Dalton Cardiovascular Research Center (M.M.-Q., F.I.R.-P., C.A.F., T.G., L.F.-S., C.M.-A., J.P., L.A.M.-L.), University of Missouri, Columbia, MO.,Department of Nutrition and Exercise Physiology (J.P.), University of Missouri, Columbia, MO
| | - Luis A Martinez-Lemus
- From the Dalton Cardiovascular Research Center (M.M.-Q., F.I.R.-P., C.A.F., T.G., L.F.-S., C.M.-A., J.P., L.A.M.-L.), University of Missouri, Columbia, MO.,Department of Biological Engineering (F.I.R.-P., L.A.M.-L.), University of Missouri, Columbia, MO.,Department of Medical Pharmacology and Physiology (L.A.M.-L.), University of Missouri, Columbia, MO
| |
Collapse
|
24
|
Li B, Yu Q, Wang R, Gratzke C, Wang X, Spek A, Herlemann A, Tamalunas A, Strittmatter F, Waidelich R, Stief CG, Hennenberg M. Inhibition of Female and Male Human Detrusor Smooth Muscle Contraction by the Rac Inhibitors EHT1864 and NSC23766. Front Pharmacol 2020; 11:409. [PMID: 32317972 PMCID: PMC7154109 DOI: 10.3389/fphar.2020.00409] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 03/18/2020] [Indexed: 12/14/2022] Open
Abstract
Introduction Lower urinary tract symptoms (LUTS) due to overactive bladder (OAB) are caused by spontaneous detrusor contractions. Medical treatment with muscarinic receptor antagonists or β3-adrenoceptor agonists aims to inhibit detrusor contractions, but overall results are unsatisfactory. Consequently, improved understanding of bladder smooth muscle contraction and identification of novel compounds for its inhibition are needed to develop alternative options. A role of the GTPase Rac1 for smooth muscle contraction has been reported from the prostate, but is unknown in the human detrusor. Here, we examined effects of the Rac inhibitors NSC23766, which may also antagonize muscarinic receptors, and EHT1864 on contraction of human detrusor tissues. Methods Female and male human detrusor tissues were obtained from radical cystectomy. Effects of NSC23766 (100 µM) and EHT1864 (100 µM) on detrusor contractions were studied in an organ bath. Results Electric field stimulation induced frequency-dependent contractions of detrusor tissues, which were inhibited by NSC23766 and EHT1864. Carbachol induced concentration-dependent contractions. Concentration response curves for carbachol were shifted to the right by NSC23766, reflected by increased EC50 values, but unchanged Emax values. EHT1864 reduced carbachol-induced contractions, resulting in reduced Emax values for carbachol. The thromboxane analog U46619 induced concentration-dependent contractions, which remained unchanged by NSC23766, but were reduced by EHT1864. Conclusions NSC23766 and EHT1864 inhibit female and male human detrusor contractions. NSC23766, but not EHT1864 competitively antagonizes muscarinic receptors. In addition to neurogenic and cholinergic contractions, EHT1864 inhibits thromboxane A2-induced detrusor contractions. The latter may be promising, as the origin of spontaneous detrusor contractions in OAB is noncholinergic. In vivo, both compounds may improve OAB-related LUTS.
Collapse
Affiliation(s)
- Bingsheng Li
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | - Qingfeng Yu
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | - Ruixiao Wang
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | - Christian Gratzke
- Department of Urology, University Hospital, LMU Munich, Munich, Germany.,Department of Urology, University of Freiburg, Freiburg, Germany
| | - Xiaolong Wang
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | - Annabel Spek
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | - Annika Herlemann
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | | | | | | | - Christian G Stief
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | - Martin Hennenberg
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
25
|
Lai WF, Wong WT. Roles of the actin cytoskeleton in aging and age-associated diseases. Ageing Res Rev 2020; 58:101021. [PMID: 31968269 DOI: 10.1016/j.arr.2020.101021] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 01/06/2020] [Accepted: 01/17/2020] [Indexed: 12/12/2022]
Abstract
The integrity of the cytoskeleton is essential to diverse cellular processes such as phagocytosis and intracellular trafficking. Disruption of the organization and dynamics of the actin cytoskeleton leads to age-associated symptoms and diseases, ranging from cancer to neurodegeneration. In addition, changes in the integrity of the actin cytoskeleton disrupt the functioning of not only somatic and stem cells but also gametes, resulting in aberrant embryonic development. Strategies to preserve the integrity and dynamics of the cytoskeleton are, therefore, potentially therapeutic to age-related disorders. The objective of this article is to revisit the current understanding of the roles played by the actin cytoskeleton in aging, and to review the opportunities and challenges for the transition of basic research into intervention development. It is hoped that, with the snapshot of evidence regarding changes in actin dynamics with advanced age, insights into future research directions can be attained.
Collapse
Affiliation(s)
- Wing-Fu Lai
- School of Pharmaceutical Sciences, Shenzhen University, PR China; School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, PR China; Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hong Kong Special Administrative Region, PR China.
| | - Wing-Tak Wong
- Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hong Kong Special Administrative Region, PR China
| |
Collapse
|
26
|
Touyz RM, Alves-Lopes R, Rios FJ, Camargo LL, Anagnostopoulou A, Arner A, Montezano AC. Vascular smooth muscle contraction in hypertension. Cardiovasc Res 2019; 114:529-539. [PMID: 29394331 PMCID: PMC5852517 DOI: 10.1093/cvr/cvy023] [Citation(s) in RCA: 433] [Impact Index Per Article: 72.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 01/30/2018] [Indexed: 12/19/2022] Open
Abstract
Hypertension is a major risk factor for many common chronic diseases, such as heart failure, myocardial infarction, stroke, vascular dementia, and chronic kidney disease. Pathophysiological mechanisms contributing to the development of hypertension include increased vascular resistance, determined in large part by reduced vascular diameter due to increased vascular contraction and arterial remodelling. These processes are regulated by complex-interacting systems such as the renin-angiotensin-aldosterone system, sympathetic nervous system, immune activation, and oxidative stress, which influence vascular smooth muscle function. Vascular smooth muscle cells are highly plastic and in pathological conditions undergo phenotypic changes from a contractile to a proliferative state. Vascular smooth muscle contraction is triggered by an increase in intracellular free calcium concentration ([Ca2+]i), promoting actin–myosin cross-bridge formation. Growing evidence indicates that contraction is also regulated by calcium-independent mechanisms involving RhoA-Rho kinase, protein Kinase C and mitogen-activated protein kinase signalling, reactive oxygen species, and reorganization of the actin cytoskeleton. Activation of immune/inflammatory pathways and non-coding RNAs are also emerging as important regulators of vascular function. Vascular smooth muscle cell [Ca2+]i not only determines the contractile state but also influences activity of many calcium-dependent transcription factors and proteins thereby impacting the cellular phenotype and function. Perturbations in vascular smooth muscle cell signalling and altered function influence vascular reactivity and tone, important determinants of vascular resistance and blood pressure. Here, we discuss mechanisms regulating vascular reactivity and contraction in physiological and pathophysiological conditions and highlight some new advances in the field, focusing specifically on hypertension.
Collapse
Affiliation(s)
- Rhian M Touyz
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Rheure Alves-Lopes
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Francisco J Rios
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Livia L Camargo
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Aikaterini Anagnostopoulou
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Anders Arner
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Augusto C Montezano
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| |
Collapse
|
27
|
Wenceslau CF, McCarthy CG, Szasz T, Calmasini FB, Mamenko M, Webb RC. Formyl peptide receptor-1 activation exerts a critical role for the dynamic plasticity of arteries via actin polymerization. Pharmacol Res 2019; 141:276-290. [PMID: 30639374 DOI: 10.1016/j.phrs.2019.01.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/07/2019] [Accepted: 01/08/2019] [Indexed: 12/20/2022]
Abstract
Several human diseases, include cancer and stroke are characterized by changes in immune system activation and vascular contractility. However, the mechanistic foundation of a vascular immuno-physiology network is still largely unknown. Formyl peptide receptor-1 (FPR-1), which plays a vital role in the function of the innate immune system, is widely expressed in arteries, but its role in vascular plasticity is unclear. We questioned why a receptor that is crucial for immune defense, and cell motility in leukocytes, would be expressed in vascular smooth muscle cells (VSMCs). We hypothesized that activation of FPR-1 in arteries is important for the temporal reorganization of actin filaments, and consequently, changes in vascular function, similar to what is observed in neutrophils. To address our hypothesis, we used FPR-1 knockout and VSMCs lacking FPR-1. We observed that FPR-1 activation induces actin polymerization in wild type VSMCs. Absence of FPR-1 in the vasculature significantly decreased vascular contraction and induced loss of myogenic tone to elevated intraluminal pressures via disruption of actin polymerization. Actin polymerization activator ameliorated these responses. In conclusion, we have established a novel role for FPR-1 in VSMC contractility and motility, similar to the one observed in sentinel cells of the innate immune system. This discovery is fundamental for vascular immuno-pathophysiology, given that FPR-1 in VSMCs not only functions as an immune system receptor, but it also has an important role for the dynamic plasticity of arteries.
Collapse
Affiliation(s)
- Camilla F Wenceslau
- Department of Physiology and Pharmacology, University of Toledo College of Medicine & Life Sciences, Toledo, OH, USA; Department of Physiology, Augusta University, Augusta, GA, USA.
| | - Cameron G McCarthy
- Department of Physiology and Pharmacology, University of Toledo College of Medicine & Life Sciences, Toledo, OH, USA; Department of Physiology, Augusta University, Augusta, GA, USA
| | - Theodora Szasz
- Department of Physiology, Augusta University, Augusta, GA, USA
| | | | - Mykola Mamenko
- Department of Physiology, Augusta University, Augusta, GA, USA
| | - R Clinton Webb
- Department of Physiology, Augusta University, Augusta, GA, USA
| |
Collapse
|
28
|
Martinez-Quinones P, McCarthy CG, Watts SW, Klee NS, Komic A, Calmasini FB, Priviero F, Warner A, Chenghao Y, Wenceslau CF. Hypertension Induced Morphological and Physiological Changes in Cells of the Arterial Wall. Am J Hypertens 2018; 31:1067-1078. [PMID: 29788246 DOI: 10.1093/ajh/hpy083] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 05/14/2018] [Indexed: 12/17/2022] Open
Abstract
Morphological and physiological changes in the vasculature have been described in the evolution and maintenance of hypertension. Hypertension-induced vascular dysfunction may present itself as a contributing, or consequential factor, to vascular remodeling caused by chronically elevated systemic arterial blood pressure. Changes in all vessel layers, from the endothelium to the perivascular adipose tissue (PVAT), have been described. This mini-review focuses on the current knowledge of the structure and function of the vessel layers, specifically muscular arteries: intima, media, adventitia, PVAT, and the cell types harbored within each vessel layer. The contributions of each cell type to vessel homeostasis and pathophysiological development of hypertension will be highlighted.
Collapse
Affiliation(s)
- Patricia Martinez-Quinones
- Department of Surgery, Medical College of Georgia at Augusta University, Augusta, Georgia
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Cameron G McCarthy
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Stephanie W Watts
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, USA
| | - Nicole S Klee
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Amel Komic
- Department of Surgery, Medical College of Georgia at Augusta University, Augusta, Georgia
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Fabiano B Calmasini
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Fernanda Priviero
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Alexander Warner
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Yu Chenghao
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Camilla F Wenceslau
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| |
Collapse
|
29
|
Ye BH, Kim EJ, Baek SE, Choi YW, Park SY, Kim CD. α-Isocubebene modulates vascular tone by inhibiting myosin light chain phosphorylation in murine thoracic aorta. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2018; 22:437-445. [PMID: 29962858 PMCID: PMC6019879 DOI: 10.4196/kjpp.2018.22.4.437] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 04/23/2018] [Accepted: 04/24/2018] [Indexed: 02/07/2023]
Abstract
α-Iso-cubebene (ICB) is a dibenzocyclooctadiene lignin contained in Schisandra chinensis (SC), a well-known medicinal herb that ameliorates cardiovascular symptoms, but the mechanism responsible for this activity has not been determined. To determine the role played by ICB on the regulation of vascular tone, we investigated the inhibitory effects of ICB on vascular contractile responses by adrenergic α-receptor agonists. In addition, we investigated the role on myosin light chain (MLC) phosphorylation and cytosolic calcium concentration in vascular smooth muscle cells (VSMC). In aortic rings isolated from C57BL/6J mice, ICB significantly attenuated the contraction induced by phenylephrine (PE) and norepinephrine (NE), whereas ICB had no effects on KCl (60 mM)-induced contraction. In vasculatures precontracted with PE, ICB caused marked relaxation of aortic rings with or without endothelium, suggesting a direct effect on VSMC. In cultured rat VSMC, PE or NE increased MLC phosphorylation and increased cytosolic calcium levels. Both of these effects were significantly suppressed by ICB. In conclusion, our results showed that ICB regulated vascular tone by inhibiting MLC phosphorylation and calcium flux into VSMC, and suggest that ICB has anti-hypertensive properties and therapeutic potential for cardiovascular disorders related to vascular hypertension.
Collapse
Affiliation(s)
- Byeong Hyeok Ye
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Eun Jung Kim
- Gene & Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Yangsan 50612, Korea
| | - Seung Eun Baek
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Korea.,Gene & Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Yangsan 50612, Korea
| | - Young Whan Choi
- College of Natural Resources & Life Sciences, Pusan National University, Miryang 50463, Korea
| | - So Youn Park
- Gene & Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Yangsan 50612, Korea
| | - Chi Dae Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Korea.,Gene & Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Yangsan 50612, Korea
| |
Collapse
|
30
|
Klein A, Joseph PD, Christensen VG, Jensen LJ, Jacobsen JCB. Lack of tone in mouse small mesenteric arteries leads to outward remodeling, which can be prevented by prolonged agonist-induced vasoconstriction. Am J Physiol Heart Circ Physiol 2018; 315:H644-H657. [PMID: 29775408 DOI: 10.1152/ajpheart.00111.2018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Inward remodeling of resistance vessels is an independent risk factor for cardiovascular events. Thus far, the remodeling process remains incompletely elucidated, but the activation level of the vascular smooth muscle cell appears to play a central role. Accordingly, previous data have suggested that an antagonistic and supposedly beneficial response, outward remodeling, may follow prolonged vasodilatation. The present study aimed to determine whether 1) outward remodeling follows 3 days of vessel culture without tone, 2) a similar response can be elicited in a much shorter 4-h timeframe, and, finally, 3) whether a 4-h response can be prevented or reversed by the presence of vasoconstrictors in the medium. Cannulated mouse small mesenteric arteries were organocultured for 3 days in the absence of tone, leading to outward remodeling that continued throughout the culture period. In more acute experiments in which cannulated small mesenteric arteries were maintained in physiological saline without tone for 4 h, we detected a similar outward remodeling that proceeded at a rate several times faster. In the 4-h experimental setting, continuous vasoconstriction to ~50% tone by abluminal application of UTP or norepinephrine + neuropeptide Y prevented outward remodeling but did not cause inward remodeling. Computational modeling was used to simulate and interpret these findings and to derive time constants of the remodeling processes. It is suggested that depriving resistance arteries of activation will lead to eutrophic outward remodeling, which can be prevented by vascular smooth muscle cell activation induced by prolonged vasoconstrictor exposure. NEW & NOTEWORTHY We have established an effective 4-h method for studying outward remodeling in pressurized mouse resistance vessels ex vivo and have determined conditions that block the remodeling response. This allows for investigating the subtle but clinically highly relevant phenomenon of outward remodeling while avoiding both laborious 3-day organoid culture of cannulated vessels and in vivo experiments lasting several weeks.
Collapse
Affiliation(s)
- Anika Klein
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark.,Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Philomeena Daphne Joseph
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Vibeke Grøsfjeld Christensen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Lars Jørn Jensen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Jens Christian Brings Jacobsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| |
Collapse
|
31
|
Bloksgaard M, Thorsted B, Brewer JR, De Mey JGR. Assessing Collagen and Elastin Pressure-dependent Microarchitectures in Live, Human Resistance Arteries by Label-free Fluorescence Microscopy. J Vis Exp 2018. [PMID: 29683445 DOI: 10.3791/57451] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The pathogenic contribution of resistance artery remodeling is documented in essential hypertension, diabetes and the metabolic syndrome. Investigations and development of microstructurally motivated mathematical models for understanding the mechanical properties of human resistance arteries in health and disease have the potential to aid understanding how disease and medical treatments affect the human microcirculation. To develop these mathematical models, it is essential to decipher the relationship between the mechanical and microarchitectural properties of the microvascular wall. In this work, we describe an ex vivo method for passive mechanical testing and simultaneous label-free three-dimensional imaging of the microarchitecture of elastin and collagen in the arterial wall of isolated human resistance arteries. The imaging protocol can be applied to resistance arteries of any species of interest. Image analyses are described for quantifying i) pressure-induced changes in internal elastic lamina branching angles and adventitial collagen straightness using Fiji and ii) collagen and elastin volume densities determined using Ilastik software. Preferably all mechanical and imaging measurements are performed on live, perfused arteries, however, an alternative approach using standard video-microscopy pressure myography in combination with post-fixation imaging of re-pressurized vessels is discussed. This alternative method provides users with different options for analysis approaches. The inclusion of the mechanical and imaging data in mathematical models of the arterial wall mechanics is discussed, and future development and additions to the protocol are proposed.
Collapse
Affiliation(s)
- Maria Bloksgaard
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark;
| | - Bjarne Thorsted
- Department of Biochemistry and Molecular Biology, University of Southern Denmark
| | - Jonathan R Brewer
- Department of Biochemistry and Molecular Biology, University of Southern Denmark
| | - Jo G R De Mey
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark; Department of Cardiac, Thoracic and Vascular Surgery, Odense University Hospital
| |
Collapse
|
32
|
Xu Q, Huff LP, Fujii M, Griendling KK. Redox regulation of the actin cytoskeleton and its role in the vascular system. Free Radic Biol Med 2017; 109:84-107. [PMID: 28285002 PMCID: PMC5497502 DOI: 10.1016/j.freeradbiomed.2017.03.004] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 02/17/2017] [Accepted: 03/06/2017] [Indexed: 12/17/2022]
Abstract
The actin cytoskeleton is critical for form and function of vascular cells, serving mechanical, organizational and signaling roles. Because many cytoskeletal proteins are sensitive to reactive oxygen species, redox regulation has emerged as a pivotal modulator of the actin cytoskeleton and its associated proteins. Here, we summarize work implicating oxidants in altering actin cytoskeletal proteins and focus on how these alterations affect cell migration, proliferation and contraction of vascular cells. Finally, we discuss the role of oxidative modification of the actin cytoskeleton in vivo and highlight its importance for vascular diseases.
Collapse
Affiliation(s)
- Qian Xu
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, 308a WMB, Atlanta, GA 30322, United States; Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Lauren P Huff
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, 308a WMB, Atlanta, GA 30322, United States
| | - Masakazu Fujii
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Japan
| | - Kathy K Griendling
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, 308a WMB, Atlanta, GA 30322, United States.
| |
Collapse
|
33
|
Schubert KM, Qiu J, Blodow S, Wiedenmann M, Lubomirov LT, Pfitzer G, Pohl U, Schneider H. The AMP-Related Kinase (AMPK) Induces Ca
2+
-Independent Dilation of Resistance Arteries by Interfering With Actin Filament Formation. Circ Res 2017; 121:149-161. [DOI: 10.1161/circresaha.116.309962] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 05/23/2017] [Accepted: 06/06/2017] [Indexed: 12/13/2022]
Abstract
Rationale:
Decreasing Ca
2+
sensitivity of vascular smooth muscle (VSM) allows for vasodilation without lowering of cytosolic Ca
2+
. This may be particularly important in states requiring maintained dilation, such as hypoxia. AMP-related kinase (AMPK) is an important cellular energy sensor in VSM. Regulation of Ca
2+
sensitivity usually is attributed to myosin light chain phosphatase activity, but findings in non-VSM identified changes in the actin cytoskeleton. The potential role of AMPK in this setting is widely unknown.
Objective:
To assess the influence of AMPK on the actin cytoskeleton in VSM of resistance arteries with regard to potential Ca
2+
desensitization of VSM contractile apparatus.
Methods and Results:
AMPK induced a slowly developing dilation at unchanged cytosolic Ca
2+
levels in potassium chloride–constricted intact arteries isolated from mouse mesenteric tissue. This dilation was not associated with changes in phosphorylation of myosin light chain or of myosin light chain phosphatase regulatory subunit. Using ultracentrifugation and confocal microscopy, we found that AMPK induced depolymerization of F-actin (filamentous actin). Imaging of arteries from LifeAct mice showed F-actin rarefaction in the midcellular portion of VSM. Immunoblotting revealed that this was associated with activation of the actin severing factor cofilin. Coimmunoprecipitation experiments indicated that AMPK leads to the liberation of cofilin from 14-3-3 protein.
Conclusions:
AMPK induces actin depolymerization, which reduces vascular tone and the response to vasoconstrictors. Our findings demonstrate a new role of AMPK in the control of actin cytoskeletal dynamics, potentially allowing for long-term dilation of microvessels without substantial changes in cytosolic Ca
2+
.
Collapse
Affiliation(s)
- Kai Michael Schubert
- From the Walter Brendel Centre of Experimental Medicine, Biomedical Center of LMU, Ludwig Maximilian University of Munich, Germany (K.M.S., J.Q., S.B., M.W., U.P., H.S.); Munich Cluster for Systems Neurology (SyNergy), Germany (K.M.S., S.B., U.P., H.S.); Deutsches Zentrum für Herz- Kreislauf-Forschung (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany (K.M.S., S.B., U.P., H.S.); and Institute of Vegetative Physiology, University of Cologne, Germany (L.T
| | - Jiehua Qiu
- From the Walter Brendel Centre of Experimental Medicine, Biomedical Center of LMU, Ludwig Maximilian University of Munich, Germany (K.M.S., J.Q., S.B., M.W., U.P., H.S.); Munich Cluster for Systems Neurology (SyNergy), Germany (K.M.S., S.B., U.P., H.S.); Deutsches Zentrum für Herz- Kreislauf-Forschung (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany (K.M.S., S.B., U.P., H.S.); and Institute of Vegetative Physiology, University of Cologne, Germany (L.T
| | - Stephanie Blodow
- From the Walter Brendel Centre of Experimental Medicine, Biomedical Center of LMU, Ludwig Maximilian University of Munich, Germany (K.M.S., J.Q., S.B., M.W., U.P., H.S.); Munich Cluster for Systems Neurology (SyNergy), Germany (K.M.S., S.B., U.P., H.S.); Deutsches Zentrum für Herz- Kreislauf-Forschung (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany (K.M.S., S.B., U.P., H.S.); and Institute of Vegetative Physiology, University of Cologne, Germany (L.T
| | - Margarethe Wiedenmann
- From the Walter Brendel Centre of Experimental Medicine, Biomedical Center of LMU, Ludwig Maximilian University of Munich, Germany (K.M.S., J.Q., S.B., M.W., U.P., H.S.); Munich Cluster for Systems Neurology (SyNergy), Germany (K.M.S., S.B., U.P., H.S.); Deutsches Zentrum für Herz- Kreislauf-Forschung (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany (K.M.S., S.B., U.P., H.S.); and Institute of Vegetative Physiology, University of Cologne, Germany (L.T
| | - Lubomir T. Lubomirov
- From the Walter Brendel Centre of Experimental Medicine, Biomedical Center of LMU, Ludwig Maximilian University of Munich, Germany (K.M.S., J.Q., S.B., M.W., U.P., H.S.); Munich Cluster for Systems Neurology (SyNergy), Germany (K.M.S., S.B., U.P., H.S.); Deutsches Zentrum für Herz- Kreislauf-Forschung (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany (K.M.S., S.B., U.P., H.S.); and Institute of Vegetative Physiology, University of Cologne, Germany (L.T
| | - Gabriele Pfitzer
- From the Walter Brendel Centre of Experimental Medicine, Biomedical Center of LMU, Ludwig Maximilian University of Munich, Germany (K.M.S., J.Q., S.B., M.W., U.P., H.S.); Munich Cluster for Systems Neurology (SyNergy), Germany (K.M.S., S.B., U.P., H.S.); Deutsches Zentrum für Herz- Kreislauf-Forschung (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany (K.M.S., S.B., U.P., H.S.); and Institute of Vegetative Physiology, University of Cologne, Germany (L.T
| | - Ulrich Pohl
- From the Walter Brendel Centre of Experimental Medicine, Biomedical Center of LMU, Ludwig Maximilian University of Munich, Germany (K.M.S., J.Q., S.B., M.W., U.P., H.S.); Munich Cluster for Systems Neurology (SyNergy), Germany (K.M.S., S.B., U.P., H.S.); Deutsches Zentrum für Herz- Kreislauf-Forschung (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany (K.M.S., S.B., U.P., H.S.); and Institute of Vegetative Physiology, University of Cologne, Germany (L.T
| | - Holger Schneider
- From the Walter Brendel Centre of Experimental Medicine, Biomedical Center of LMU, Ludwig Maximilian University of Munich, Germany (K.M.S., J.Q., S.B., M.W., U.P., H.S.); Munich Cluster for Systems Neurology (SyNergy), Germany (K.M.S., S.B., U.P., H.S.); Deutsches Zentrum für Herz- Kreislauf-Forschung (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany (K.M.S., S.B., U.P., H.S.); and Institute of Vegetative Physiology, University of Cologne, Germany (L.T
| |
Collapse
|
34
|
Bloksgaard M, Leurgans TM, Spronck B, Heusinkveld MHG, Thorsted B, Rosenstand K, Nissen I, Hansen UM, Brewer JR, Bagatolli LA, Rasmussen LM, Irmukhamedov A, Reesink KD, De Mey JGR. Imaging and modeling of acute pressure-induced changes of collagen and elastin microarchitectures in pig and human resistance arteries. Am J Physiol Heart Circ Physiol 2017; 313:H164-H178. [DOI: 10.1152/ajpheart.00110.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 03/30/2017] [Accepted: 04/14/2017] [Indexed: 01/15/2023]
Abstract
The impact of disease-related changes in the extracellular matrix (ECM) on the mechanical properties of human resistance arteries largely remains to be established. Resistance arteries from both pig and human parietal pericardium (PRA) display a different ECM microarchitecture compared with frequently used rodent mesenteric arteries. We hypothesized that the biaxial mechanics of PRA mirror pressure-induced changes in the ECM microarchitecture. This was tested using isolated pig PRA as a model system, integrating vital imaging, pressure myography, and mathematical modeling. Collagenase and elastase digestions were applied to evaluate the load-bearing roles of collagen and elastin, respectively. The incremental elastic modulus linearly related to the straightness of adventitial collagen fibers circumferentially and longitudinally (both R2 ≥ 0.99), whereas there was a nonlinear relationship to the internal elastic lamina elastin fiber branching angles. Mathematical modeling suggested a collagen recruitment strain (means ± SE) of 1.1 ± 0.2 circumferentially and 0.20 ± 0.01 longitudinally, corresponding to a pressure of ~40 mmHg, a finding supported by the vital imaging. The integrated method was tested on human PRA to confirm its validity. These showed limited circumferential distensibility and elongation and a collagen recruitment strain of 0.8 ± 0.1 circumferentially and 0.06 ± 0.02 longitudinally, reached at a distending pressure below 20 mmHg. This was confirmed by vital imaging showing negligible microarchitectural changes of elastin and collagen upon pressurization. In conclusion, we show here, for the first time in resistance arteries, a quantitative relationship between pressure-induced changes in the extracellular matrix and the arterial wall mechanics. The strength of the integrated methods invites for future detailed studies of microvascular pathologies. NEW & NOTEWORTHY This is the first study to quantitatively relate pressure-induced microstructural changes in resistance arteries to the mechanics of their wall. Principal findings using a pig model system were confirmed in human arteries. The combined methods provide a strong tool for future hypothesis-driven studies of microvascular pathologies.
Collapse
Affiliation(s)
- Maria Bloksgaard
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Thomas M. Leurgans
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Bart Spronck
- Department of Biomedical Engineering, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Maarten H. G. Heusinkveld
- Department of Biomedical Engineering, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Bjarne Thorsted
- MEMPHYS-Center for Biomembrane Physics, University of Southern Denmark, Odense, Denmark
| | - Kristoffer Rosenstand
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Inger Nissen
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Ulla M. Hansen
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Jonathan R. Brewer
- MEMPHYS-Center for Biomembrane Physics, University of Southern Denmark, Odense, Denmark
| | - Luis A. Bagatolli
- MEMPHYS-Center for Biomembrane Physics, University of Southern Denmark, Odense, Denmark
| | - Lars M. Rasmussen
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark; and
| | - Akhmadjon Irmukhamedov
- Department of Cardiac, Thoracic and Vascular Surgery, Odense University Hospital, Odense, Denmark
| | - Koen D. Reesink
- Department of Biomedical Engineering, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Jo G. R. De Mey
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Department of Cardiac, Thoracic and Vascular Surgery, Odense University Hospital, Odense, Denmark
| |
Collapse
|
35
|
Wang Y, Zhou Q, Wu B, Zhou H, Zhang X, Jiang W, Wang L, Wang A. Propofol induces excessive vasodilation of aortic rings by inhibiting protein kinase Cβ2 and θ in spontaneously hypertensive rats. Br J Pharmacol 2017; 174:1984-2000. [PMID: 28369981 DOI: 10.1111/bph.13797] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 02/21/2017] [Accepted: 03/19/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE Exaggerated hypotension following administration of propofol is strongly predicted in patients with hypertension. Increased PKCs play a crucial role in regulating vascular tone. We studied whether propofol induces vasodilation by inhibiting increased PKC activity in spontaneously hypertensive rats (SHRs) and, if so, whether contractile Ca2+ sensitization pathways and filamentous-globular (F/G) actin dynamics were involved. EXPERIMENTAL APPROACH Rings of thoracic aorta, denuded of endothelium, from normotensive Wistar-Kyoto (WKY) rats and SHR were prepared for functional studies. Expression and activity of PKCs in vascular smooth muscle (VSM) cells were determined by Western blot analysis and elisa respectively. Phosphorylation of the key proteins in PKC Ca2+ sensitization pathways was also examined. Actin polymerization was evaluated by differential centrifugation to probe G- and F-actin content. KEY RESULTS Basal expression and activity of PKCβ2 and PKCθ were increased in aortic VSMs of SHR, compared with those from WKY rats. Vasorelaxation of SHR aortas by propofol was markedly attenuated by LY333531 (a specific PKCβ inhibitor) or the PKCθ pseudo-substrate inhibitor. Furthermore, noradrenaline-enhanced phosphorylation, and the translocation of PKCβ2 and PKCθ, was inhibited by propofol, with decreased actin polymerization and PKCβ2-mediated Ca2+ sensitization pathway in SHR aortas. CONCLUSION AND IMPLICATIONS Propofol suppressed increased PKCβ2 and PKCθ activity, which was partly responsible for exaggerated vasodilation in SHR. This suppression results in inhibition of actin polymerization, as well as that of the PKCβ2- but not PKCθ-mediated, Ca2+ sensitization pathway. These data provide a novel explanation for the unwanted side effects of propofol.
Collapse
Affiliation(s)
- Yan Wang
- Department of Anesthesiology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Quanhong Zhou
- Department of Anesthesiology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Bin Wu
- Department of Anesthesiology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Huixuan Zhou
- Department of Anesthesiology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Xiaoli Zhang
- Department of Anesthesiology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Wei Jiang
- Department of Anesthesiology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Li Wang
- Department of Anesthesiology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Aizhong Wang
- Department of Anesthesiology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
36
|
Mottola G, Chatterjee A, Wu B, Chen M, Conte MS. Aspirin-triggered resolvin D1 attenuates PDGF-induced vascular smooth muscle cell migration via the cyclic adenosine monophosphate/protein kinase A (cAMP/PKA) pathway. PLoS One 2017; 12:e0174936. [PMID: 28362840 PMCID: PMC5376330 DOI: 10.1371/journal.pone.0174936] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 03/17/2017] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Resolvin D1 (RvD1) is a specialized pro-resolving lipid mediator that has been previously shown to attenuate vascular smooth muscle cell (VSMC) migration, a key process in the development of intimal hyperplasia. We sought to investigate the role of the cAMP/PKA pathway in mediating the effects of the aspirin-triggered epimer 17R-RvD1 (AT-RvD1) on VSMC migration. METHODS VSMCs were harvested from human saphenous veins. VSMCs were analyzed for intracellular cAMP levels and PKA activity after exposure to AT-RvD1. Platelet-derived growth factor (PDGF)-induced migration and cytoskeletal changes in VSMCs were observed through scratch, Transwell, and cell shape assays in the presence or absence of a PKA inhibitor (Rp-8-Br-cAMP). Further investigation of the pathways involved in AT-RvD1 signaling was performed by measuring Rac1 activity, vasodilator stimulated phosphoprotein (VASP) phosphorylation and paxillin translocation. Finally, we examined the role of RvD1 receptors (GPR32 and ALX/FPR2) in AT-RvD1 induced effects on VSMC migration and PKA activity. RESULTS Treatment with AT-RvD1 induced a significant increase in cAMP levels and PKA activity in VSMCs at 5 minutes and 30 minutes, respectively. AT-RvD1 attenuated PDGF-induced VSMC migration and cytoskeletal rearrangements. These effects were attenuated by the PKA inhibitor Rp-8-Br-cAMP, suggesting cAMP/PKA involvement. Treatment of VSMC with AT-RvD1 inhibited PDGF-stimulated Rac1 activity, increased VASP phosphorylation, and attenuated paxillin localization to focal adhesions; these effects were negated by the addition of Rp-8-Br-cAMP. The effects of AT-RvD1 on VSMC migration and PKA activity were attenuated by blocking ALX/FPR2, suggesting an important role of this G-protein coupled receptor. CONCLUSIONS Our results suggest that AT-RvD1 attenuates PDGF-induced VSMC migration via ALX/FPR2 and cAMP/PKA. Interference with Rac1, VASP and paxillin function appear to mediate the downstream effects of AT-RvD1 on VSMC migration.
Collapse
Affiliation(s)
- Giorgio Mottola
- Department of Surgery, Division of Vascular and Endovascular Surgery, Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, United States of America
- * E-mail:
| | - Anuran Chatterjee
- Department of Surgery, Division of Vascular and Endovascular Surgery, Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, United States of America
| | - Bian Wu
- Department of Surgery, Division of Vascular and Endovascular Surgery, Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, United States of America
| | - Mian Chen
- Department of Surgery, Division of Vascular and Endovascular Surgery, Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, United States of America
| | - Michael S. Conte
- Department of Surgery, Division of Vascular and Endovascular Surgery, Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, United States of America
| |
Collapse
|
37
|
Baron-Menguy C, Domenga-Denier V, Ghezali L, Faraci FM, Joutel A. Increased Notch3 Activity Mediates Pathological Changes in Structure of Cerebral Arteries. Hypertension 2016; 69:60-70. [PMID: 27821617 DOI: 10.1161/hypertensionaha.116.08015] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 07/07/2016] [Accepted: 08/26/2016] [Indexed: 01/24/2023]
Abstract
CADASIL (Cerebral Autosomal Dominant Arteriopathy With Subcortical Infarcts and Leukoencephalopathy), the most frequent genetic cause of stroke and vascular dementia, is caused by highly stereotyped mutations in the NOTCH3 receptor, which is predominantly expressed in vascular smooth muscle. The well-established TgNotch3R169C mouse model develops characteristic features of the human disease, with deposition of NOTCH3 and other proteins, including TIMP3 (tissue inhibitor of metalloproteinase 3), on brain vessels, as well as reduced maximal dilation, and attenuated myogenic tone of cerebral arteries, but without elevated blood pressure. Increased TIMP3 levels were recently shown to be a major determinant of altered myogenic tone. In this study, we investigated the contribution of TIMP3 and Notch3 signaling to the impairment of maximal vasodilator capacity caused by the archetypal R169C mutation. Maximally dilated cerebral arteries in TgNotch3R169C mice exhibited a decrease in lumen diameter over a range of physiological pressures that occurred before myogenic tone deficits. This defect was not prevented by genetic reduction of TIMP3 in TgNotch3R169C mice and was not observed in mice overexpressing TIMP3. Knock-in mice with the R169C mutation (Notch3R170C/R170C) exhibited similar reductions in arterial lumen, and both TgNotch3R169C and Notch3R170C/R170C mice showed increased cerebral artery expression of Notch3 target genes. Reduced maximal vasodilation was prevented by conditional reduction of Notch activity in smooth muscle of TgNotch3R169C mice and mimicked by conditional activation of Notch3 in smooth muscle, an effect that was blood pressure-independent. We conclude that increased Notch3 activity mediates reduction in maximal dilator capacity of cerebral arteries in CADASIL and may contribute to reductions in cerebral blood flow.
Collapse
Affiliation(s)
- Celine Baron-Menguy
- From the Genetics and Pathogenesis of Cerebrovascular Diseases, INSERM, UMRS 1161, and Univ Paris Diderot, Sorbonne Paris Cité, Paris, France (C.B.-M., V.D.-D., L.G., A.J.); DHU NeuroVasc Sorbonne Paris Cité, Paris, France (C.B.-M., V.D.-D., L.G., A.J.); and Departments of Internal Medicine and Pharmacology, Francois M. Abboud Cardiovascular Center, The University of Iowa Carver College of Medicine, Iowa City Veterans Affairs Healthcare System (F.M.F.)
| | - Valérie Domenga-Denier
- From the Genetics and Pathogenesis of Cerebrovascular Diseases, INSERM, UMRS 1161, and Univ Paris Diderot, Sorbonne Paris Cité, Paris, France (C.B.-M., V.D.-D., L.G., A.J.); DHU NeuroVasc Sorbonne Paris Cité, Paris, France (C.B.-M., V.D.-D., L.G., A.J.); and Departments of Internal Medicine and Pharmacology, Francois M. Abboud Cardiovascular Center, The University of Iowa Carver College of Medicine, Iowa City Veterans Affairs Healthcare System (F.M.F.)
| | - Lamia Ghezali
- From the Genetics and Pathogenesis of Cerebrovascular Diseases, INSERM, UMRS 1161, and Univ Paris Diderot, Sorbonne Paris Cité, Paris, France (C.B.-M., V.D.-D., L.G., A.J.); DHU NeuroVasc Sorbonne Paris Cité, Paris, France (C.B.-M., V.D.-D., L.G., A.J.); and Departments of Internal Medicine and Pharmacology, Francois M. Abboud Cardiovascular Center, The University of Iowa Carver College of Medicine, Iowa City Veterans Affairs Healthcare System (F.M.F.)
| | - Frank M Faraci
- From the Genetics and Pathogenesis of Cerebrovascular Diseases, INSERM, UMRS 1161, and Univ Paris Diderot, Sorbonne Paris Cité, Paris, France (C.B.-M., V.D.-D., L.G., A.J.); DHU NeuroVasc Sorbonne Paris Cité, Paris, France (C.B.-M., V.D.-D., L.G., A.J.); and Departments of Internal Medicine and Pharmacology, Francois M. Abboud Cardiovascular Center, The University of Iowa Carver College of Medicine, Iowa City Veterans Affairs Healthcare System (F.M.F.)
| | - Anne Joutel
- From the Genetics and Pathogenesis of Cerebrovascular Diseases, INSERM, UMRS 1161, and Univ Paris Diderot, Sorbonne Paris Cité, Paris, France (C.B.-M., V.D.-D., L.G., A.J.); DHU NeuroVasc Sorbonne Paris Cité, Paris, France (C.B.-M., V.D.-D., L.G., A.J.); and Departments of Internal Medicine and Pharmacology, Francois M. Abboud Cardiovascular Center, The University of Iowa Carver College of Medicine, Iowa City Veterans Affairs Healthcare System (F.M.F.).
| |
Collapse
|
38
|
Prasad AM, Ketsawatsomkron P, Nuno DW, Koval OM, Dibbern ME, Venema AN, Sigmund CD, Lamping KG, Grumbach IM. Role of CaMKII in Ang-II-dependent small artery remodeling. Vascul Pharmacol 2016; 87:172-179. [PMID: 27658984 DOI: 10.1016/j.vph.2016.09.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 08/22/2016] [Accepted: 09/18/2016] [Indexed: 01/21/2023]
Abstract
Angiotensin-II (Ang-II) is a well-established mediator of vascular remodeling. The multifunctional calcium-calmodulin-dependent kinase II (CaMKII) is activated by Ang-II and regulates Erk1/2 and Akt-dependent signaling in cultured smooth muscle cells in vitro. Its role in Ang-II-dependent vascular remodeling in vivo is far less defined. Using a model of transgenic CaMKII inhibition selectively in smooth muscle cells, we found that CaMKII inhibition exaggerated remodeling after chronic Ang-II treatment and agonist-dependent vasoconstriction in second-order mesenteric arteries. These findings were associated with increased mRNA and protein expression of smooth muscle structural proteins. As a potential mechanism, CaMKII reduced serum response factor-dependent transcriptional activity. In summary, our findings identify CaMKII as an important regulator of smooth muscle function in Ang-II hypertension in vivo.
Collapse
Affiliation(s)
- Anand M Prasad
- Department of Medicine, Carver College, University of Iowa, Iowa City, United States
| | - Pimonrat Ketsawatsomkron
- Department of Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, United States
| | - Daniel W Nuno
- Department of Medicine, Carver College, University of Iowa, Iowa City, United States; Department of Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, United States
| | - Olha M Koval
- Department of Medicine, Carver College, University of Iowa, Iowa City, United States
| | - Megan E Dibbern
- Department of Medicine, Carver College, University of Iowa, Iowa City, United States
| | - Ashlee N Venema
- Department of Medicine, Carver College, University of Iowa, Iowa City, United States
| | - Curt D Sigmund
- Department of Medicine, Carver College, University of Iowa, Iowa City, United States; Department of Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, United States; Department of Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa, Iowa City, United States
| | - Kathryn G Lamping
- Department of Medicine, Carver College, University of Iowa, Iowa City, United States; Department of Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, United States; Iowa City VA Healthcare System, Iowa City, United States
| | - Isabella M Grumbach
- Department of Medicine, Carver College, University of Iowa, Iowa City, United States; Iowa City VA Healthcare System, Iowa City, United States.
| |
Collapse
|
39
|
Freeman BM, Univers J, Fisher RK, Kirkpatrick SS, Klein FA, Freeman MB, Mountain DJH, Grandas OH. Testosterone replacement attenuates intimal hyperplasia development in an androgen deficient model of vascular injury. J Surg Res 2016; 207:53-62. [PMID: 27979488 DOI: 10.1016/j.jss.2016.08.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 07/04/2016] [Accepted: 08/03/2016] [Indexed: 12/31/2022]
Abstract
BACKGROUND Androgen deficiency (AD) is associated with increased risk of vascular disease. Dysfunctional remodeling of the vessel wall and atypical proliferative potential of vascular smooth muscle cells (VSMCs) are fundamental processes in the development of intimal hyperplasia (IH). We have demonstrated an inverse relationship between dihydrotestosterone (DHT) levels, matrix metalloproteinase activity, and VSMC migration and proliferation in vitro. Here, we investigated the role of AD and testosterone (TST) replacement in IH development in an animal model of vascular injury to elucidate mechanisms modulated by AD that could be playing a role in the development of vascular pathogenesis. METHODS Aged orchiectomized male rats underwent TST supplementation via controlled release pellet (0.5-35 mg). Young adult and middle-age adult intact (MI) and orchiectomized placebo (Plac) groups served as controls. All groups underwent balloon angioplasty of the left common carotid at a 14-d post-TST. Carotid tissue was collected at a 14-d post-balloon angioplasty and subjected to morphologic and immunohistochemical analyses. Human male VSMCs were treated with DHT (0-3000 nM) for 24 h then subjected to quantitative PCR for gene expression analyses and costained for F-actin and G-actin for visualization of cytoskeletal organization. RESULTS I:M ratio was increased in Plac, subphysiological, low-physiological, and high pharmacologic level TST animals compared with MI controls but was decreased with high-physiological TST supplementation. Injury-induced expression of previously defined matrix metalloproteinase remodeling enzymes was not significantly affected by TST status. Urotensin (UTS) receptor (UTSR) staining was low in injured vessels of all young adult intact, MI, and Plac controls but was significantly upregulated in all groups receiving exogenous TST supplementation, irrespective of dose. In vitro DHT exposure increased the expression of UTSR in VSMCs in a dose-dependent manner. However, this did not correlate with any change in proliferative markers. F:G actin staining revealed that DHT-induced cytoskeletal organization in a dose-dependent manner. CONCLUSIONS AD increased IH development in response to vascular injury, whereas physiological TST replacement attenuated this effect. AD-induced IH occurs independent of matrix remodeling mechanisms known to be heavily involved in vascular dysfunction, and AD alone does not affect the UTS and/or UTSR mechanism. Exogenous TST and/or DHT increases UTSR pathway signaling in vitro and in vivo. This modulation correlates to a shift in cytoskeletal organization and may exacerbate vasoconstrictive pathogenesis. While physiological TST replacement attenuates AD-modulated IH development, its UTS-mediated effect on vasotone may prove deleterious to overall vascular function.
Collapse
Affiliation(s)
- Brian M Freeman
- Department of Surgery, University of Tennessee Graduate School of Medicine, Knoxville, Tennessee
| | - Junior Univers
- Department of Surgery, University of Tennessee Graduate School of Medicine, Knoxville, Tennessee
| | - Richard K Fisher
- Department of Surgery, University of Tennessee Graduate School of Medicine, Knoxville, Tennessee
| | - Stacy S Kirkpatrick
- Department of Surgery, University of Tennessee Graduate School of Medicine, Knoxville, Tennessee
| | - Frederick A Klein
- Department of Surgery, University of Tennessee Graduate School of Medicine, Knoxville, Tennessee
| | - Michael B Freeman
- Department of Surgery, University of Tennessee Graduate School of Medicine, Knoxville, Tennessee
| | - Deidra J H Mountain
- Department of Surgery, University of Tennessee Graduate School of Medicine, Knoxville, Tennessee
| | - Oscar H Grandas
- Department of Surgery, University of Tennessee Graduate School of Medicine, Knoxville, Tennessee.
| |
Collapse
|
40
|
Chen YC, Yuan TY, Zhang HF, Wang DS, Niu ZR, Li L, Fang LH, Du GH. Fasudil evokes vasodilatation of rat mesenteric vascular bed via Ca(2+) channels and Rho/ROCK pathway. Eur J Pharmacol 2016; 788:226-233. [PMID: 27346833 DOI: 10.1016/j.ejphar.2016.06.041] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 06/21/2016] [Accepted: 06/22/2016] [Indexed: 01/24/2023]
Abstract
As a Rho kinase (ROCK) inhibitor, fasudil has been used in clinical trials of several cardiovascular diseases. This study was to investigate the vasorelaxant effect of fasudil on resistance arterial rings including mesenteric, renal, ventral tail and basilar artery. We also examined the potential mechanisms of its vasodilatory action using mesenteric artery rings. A DMT multiwire myograph system was used to test the tension of isolated small arteries. K(+) channel blockers, NO-cGMP pathway blockers and Ca(2+)-free physiological salt solution (PSS) were employed to verify the underlying mechanisms. Fasudil (10(-7)-10(-4)M) relaxed four types of small artery rings pre-contracted by 60mmol/l KCl (pEC50: 6.01±0.09, 5.47±0.03, 5.54±0.04, and 5.72±0.10 for mesenteric, renal, ventral tail and basilar artery rings, respectively). Pre-incubation with fasudil (1, 3, or 10μmol/l) attenuated KCl (10-60mmol/l) and angiotensin II (Ang II; 1μmol/l)-induced vasoconstriction in mesenteric artery rings. Fasudil at the concentration of 10(-6)mol/l showed different relaxant potency in endothelium intact (pEC50:6.01±0.09) or denued (5.75±0.06) mesenteric artery. The influx and release of Ca(2+) were inhibited by fasudil. In addition, fasudil could block the increased phosphorylation level of myosin light chain (MLC) and myosin-binding subunit of myosin phosphatase (MYPT1) induced by Ang II. However, pretreatment with various K(+) channel blockers did not affect the relaxant effects of fasudil remarkably. The present results demonstrate that fasudil has a vasorelaxant effect on isolated rat resistance arteries, including mesenteric, renal, ventral tail and basilar artery, and may exert its action through the endothelium, Ca(2+) channels, and the Rho/ROCK pathway.
Collapse
Affiliation(s)
- Yu-Cai Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, China
| | - Tian-Yi Yuan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, China
| | - Hui-Fang Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, China
| | - Dan-Shu Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, China
| | - Zi-Ran Niu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, China
| | - Li Li
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Lian-Hua Fang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, China.
| | - Guan-Hua Du
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
41
|
Pennington KA, Ramirez-Perez FI, Pollock KE, Talton OO, Foote CA, Reyes-Aldasoro CC, Wu HH, Ji T, Martinez-Lemus LA, Schulz LC. Maternal Hyperleptinemia Is Associated with Male Offspring's Altered Vascular Function and Structure in Mice. PLoS One 2016; 11:e0155377. [PMID: 27187080 PMCID: PMC4871503 DOI: 10.1371/journal.pone.0155377] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 04/27/2016] [Indexed: 12/27/2022] Open
Abstract
Children of mothers with gestational diabetes have greater risk of developing hypertension but little is known about the mechanisms by which this occurs. The objective of this study was to test the hypothesis that high maternal concentrations of leptin during pregnancy, which are present in mothers with gestational diabetes and/or obesity, alter blood pressure, vascular structure and vascular function in offspring. Wildtype (WT) offspring of hyperleptinemic, normoglycemic, Leprdb/+ dams were compared to genotype matched offspring of WT-control dams. Vascular function was assessed in male offspring at 6, and at 31 weeks of age after half the offspring had been fed a high fat, high sucrose diet (HFD) for 6 weeks. Blood pressure was increased by HFD but not affected by maternal hyperleptinemia. On a standard diet, offspring of hyperleptinemic dams had outwardly remodeled mesenteric arteries and an enhanced vasodilatory response to insulin. In offspring of WT but not Leprdb/+ dams, HFD induced vessel hypertrophy and enhanced vasodilatory responses to acetylcholine, while HFD reduced insulin responsiveness in offspring of hyperleptinemic dams. Offspring of hyperleptinemic dams had stiffer arteries regardless of diet. Therefore, while maternal hyperleptinemia was largely beneficial to offspring vascular health under a standard diet, it had detrimental effects in offspring fed HFD. These results suggest that circulating maternal leptin concentrations may interact with other factors in the pre- and post -natal environments to contribute to altered vascular function in offspring of diabetic pregnancies.
Collapse
Affiliation(s)
- Kathleen A. Pennington
- Department of Obstetrics, Gynecology, and Women’s Health, University of Missouri, Columbia, Missouri, United States of America
| | - Francisco I. Ramirez-Perez
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States of America
- Department of Biological Engineering, University of Missouri, Columbia, Missouri, United States of America
| | - Kelly E. Pollock
- Department of Obstetrics, Gynecology, and Women’s Health, University of Missouri, Columbia, Missouri, United States of America
| | - Omonseigho O. Talton
- Department of Obstetrics, Gynecology, and Women’s Health, University of Missouri, Columbia, Missouri, United States of America
- Division of Biological Sciences, University of Missouri, Columbia, Missouri, United States of America
| | - Christopher A. Foote
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States of America
| | | | - Ho-Hsiang Wu
- Department of Statistics, University of Missouri, Columbia, Missouri, United States of America
| | - Tieming Ji
- Department of Statistics, University of Missouri, Columbia, Missouri, United States of America
| | - Luis A. Martinez-Lemus
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States of America
- Department of Biological Engineering, University of Missouri, Columbia, Missouri, United States of America
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, United States of America
- * E-mail: (LAM); (LCS)
| | - Laura C. Schulz
- Department of Obstetrics, Gynecology, and Women’s Health, University of Missouri, Columbia, Missouri, United States of America
- Division of Biological Sciences, University of Missouri, Columbia, Missouri, United States of America
- * E-mail: (LAM); (LCS)
| |
Collapse
|
42
|
MISÁRKOVÁ E, BEHULIAK M, BENCZE M, ZICHA J. Excitation-Contraction Coupling and Excitation-Transcription Coupling in Blood Vessels: Their Possible Interactions in Hypertensive Vascular Remodeling. Physiol Res 2016; 65:173-91. [DOI: 10.33549/physiolres.933317] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Vascular smooth muscle cells (VSMC) display considerable phenotype plasticity which can be studied in vivo on vascular remodeling which occurs during acute or chronic vascular injury. In differentiated cells, which represent contractile phenotype, there are characteristic rapid transient changes of intracellular Ca2+ concentration ([Ca2+]i), while the resting cytosolic [Ca2+]i concentration is low. It is mainly caused by two components of the Ca2+ signaling pathways: Ca2+ entry via L-type voltage-dependent Ca2+ channels and dynamic involvement of intracellular stores. Proliferative VSMC phenotype is characterized by long-lasting [Ca2+]i oscillations accompanied by sustained elevation of basal [Ca2+]i. During the switch from contractile to proliferative phenotype there is a general transition from voltage-dependent Ca2+ entry to voltage-independent Ca2+ entry into the cell. These changes are due to the altered gene expression which is dependent on specific transcription factors activated by various stimuli. It is an open question whether abnormal VSMC phenotype reported in rats with genetic hypertension (such as spontaneously hypertensive rats) might be partially caused by a shift from contractile to proliferative VSMC phenotype.
Collapse
Affiliation(s)
| | | | | | - J. ZICHA
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
43
|
Brozovich FV, Nicholson CJ, Degen CV, Gao YZ, Aggarwal M, Morgan KG. Mechanisms of Vascular Smooth Muscle Contraction and the Basis for Pharmacologic Treatment of Smooth Muscle Disorders. Pharmacol Rev 2016; 68:476-532. [PMID: 27037223 PMCID: PMC4819215 DOI: 10.1124/pr.115.010652] [Citation(s) in RCA: 344] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The smooth muscle cell directly drives the contraction of the vascular wall and hence regulates the size of the blood vessel lumen. We review here the current understanding of the molecular mechanisms by which agonists, therapeutics, and diseases regulate contractility of the vascular smooth muscle cell and we place this within the context of whole body function. We also discuss the implications for personalized medicine and highlight specific potential target molecules that may provide opportunities for the future development of new therapeutics to regulate vascular function.
Collapse
Affiliation(s)
- F V Brozovich
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| | - C J Nicholson
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| | - C V Degen
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| | - Yuan Z Gao
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| | - M Aggarwal
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| | - K G Morgan
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| |
Collapse
|
44
|
Hien TT, Turczyńska KM, Dahan D, Ekman M, Grossi M, Sjögren J, Nilsson J, Braun T, Boettger T, Garcia-Vaz E, Stenkula K, Swärd K, Gomez MF, Albinsson S. Elevated Glucose Levels Promote Contractile and Cytoskeletal Gene Expression in Vascular Smooth Muscle via Rho/Protein Kinase C and Actin Polymerization. J Biol Chem 2016; 291:3552-68. [PMID: 26683376 PMCID: PMC4751395 DOI: 10.1074/jbc.m115.654384] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 12/17/2015] [Indexed: 12/22/2022] Open
Abstract
Both type 1 and type 2 diabetes are associated with increased risk of cardiovascular disease. This is in part attributed to the effects of hyperglycemia on vascular endothelial and smooth muscle cells, but the underlying mechanisms are not fully understood. In diabetic animal models, hyperglycemia results in hypercontractility of vascular smooth muscle possibly due to increased activation of Rho-kinase. The aim of the present study was to investigate the regulation of contractile smooth muscle markers by glucose and to determine the signaling pathways that are activated by hyperglycemia in smooth muscle cells. Microarray, quantitative PCR, and Western blot analyses revealed that both mRNA and protein expression of contractile smooth muscle markers were increased in isolated smooth muscle cells cultured under high compared with low glucose conditions. This effect was also observed in hyperglycemic Akita mice and in diabetic patients. Elevated glucose activated the protein kinase C and Rho/Rho-kinase signaling pathways and stimulated actin polymerization. Glucose-induced expression of contractile smooth muscle markers in cultured cells could be partially or completely repressed by inhibitors of advanced glycation end products, L-type calcium channels, protein kinase C, Rho-kinase, actin polymerization, and myocardin-related transcription factors. Furthermore, genetic ablation of the miR-143/145 cluster prevented the effects of glucose on smooth muscle marker expression. In conclusion, these data demonstrate a possible link between hyperglycemia and vascular disease states associated with smooth muscle contractility.
Collapse
MESH Headings
- Actin Cytoskeleton/metabolism
- Actin Cytoskeleton/pathology
- Aged
- Animals
- Atherosclerosis/enzymology
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Cells, Cultured
- Contractile Proteins/agonists
- Contractile Proteins/genetics
- Contractile Proteins/metabolism
- Cytoskeletal Proteins/agonists
- Cytoskeletal Proteins/genetics
- Cytoskeletal Proteins/metabolism
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 2/complications
- Diabetic Angiopathies/enzymology
- Diabetic Angiopathies/metabolism
- Diabetic Angiopathies/pathology
- Gene Expression Regulation
- Humans
- Male
- Mice, Knockout
- Mice, Mutant Strains
- MicroRNAs/metabolism
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Protein Kinase C/chemistry
- Protein Kinase C/metabolism
- Signal Transduction
- rho GTP-Binding Proteins/agonists
- rho GTP-Binding Proteins/metabolism
- rho-Associated Kinases/chemistry
- rho-Associated Kinases/metabolism
Collapse
Affiliation(s)
- Tran Thi Hien
- From the Departments of Experimental Medical Sciences and
| | | | - Diana Dahan
- From the Departments of Experimental Medical Sciences and
| | - Mari Ekman
- From the Departments of Experimental Medical Sciences and
| | - Mario Grossi
- From the Departments of Experimental Medical Sciences and
| | - Johan Sjögren
- Clinical Sciences, Lund University, BMC D12, SE-221 84 Lund, Sweden and
| | - Johan Nilsson
- Clinical Sciences, Lund University, BMC D12, SE-221 84 Lund, Sweden and
| | - Thomas Braun
- the Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany, and
| | - Thomas Boettger
- the Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany, and
| | - Eliana Garcia-Vaz
- the Department of Clinical Sciences in Malmö, Lund University, 205 02 Malmö, Sweden
| | - Karin Stenkula
- From the Departments of Experimental Medical Sciences and
| | - Karl Swärd
- From the Departments of Experimental Medical Sciences and
| | - Maria F Gomez
- the Department of Clinical Sciences in Malmö, Lund University, 205 02 Malmö, Sweden
| | | |
Collapse
|
45
|
Little R, Cartwright EJ, Neyses L, Austin C. Plasma membrane calcium ATPases (PMCAs) as potential targets for the treatment of essential hypertension. Pharmacol Ther 2016; 159:23-34. [PMID: 26820758 DOI: 10.1016/j.pharmthera.2016.01.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The incidence of hypertension, the major modifiable risk factor for cardiovascular disease, is increasing. Thus, there is a pressing need for the development of new and more effective strategies to prevent and treat hypertension. Development of these relies on a continued evolution of our understanding of the mechanisms which control blood pressure (BP). Resistance arteries are important in the regulation of total peripheral resistance and BP; changes in their structure and function are strongly associated with hypertension. Anti-hypertensives which both reduce BP and reverse changes in resistance arterial structure reduce cardiovascular risk more than therapies which reduce BP alone. Hence, identification of novel potential vascular targets which modify BP is important. Hypertension is a multifactorial disorder which may include a genetic component. Genome wide association studies have identified ATP2B1, encoding the calcium pump plasma membrane calcium ATPase 1 (PMCA1), as having a strong association with BP and hypertension. Knockdown or reduced PMCA1 expression in mice has confirmed a physiological role for PMCA1 in BP and resistance arterial regulation. Altered expression or inhibition of PMCA4 has also been shown to modulate these parameters. The mechanisms whereby PMCA1 and 4 can modulate vascular function remain to be fully elucidated but may involve regulation of intracellular calcium homeostasis and/or comprise a structural role. However, clear physiological links between PMCA and BP, coupled with experimental studies directly linking PMCA1 and 4 to changes in BP and arterial function, suggest that they may be important targets for the development of new pharmacological modulators of BP.
Collapse
Affiliation(s)
- Robert Little
- The Institute of Cardiovascular Sciences, The University of Manchester, UK
| | | | - Ludwig Neyses
- The Institute of Cardiovascular Sciences, The University of Manchester, UK
| | - Clare Austin
- Faculty of Health and Social Care, Edge Hill University, UK.
| |
Collapse
|
46
|
Foote CA, Castorena-Gonzalez JA, Staiculescu MC, Clifford PS, Hill MA, Meininger GA, Martinez-Lemus LA. Brief serotonin exposure initiates arteriolar inward remodeling processes in vivo that involve transglutaminase activation and actin cytoskeleton reorganization. Am J Physiol Heart Circ Physiol 2015; 310:H188-98. [PMID: 26566730 DOI: 10.1152/ajpheart.00666.2015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 11/10/2015] [Indexed: 12/30/2022]
Abstract
Inward remodeling of the resistance vasculature is strongly associated with life-threatening cardiovascular events. Previous studies have demonstrated that both actin polymerization and the activation of transglutaminases mediate early stages of the transition from a structurally normal vessel to an inwardly remodeled one. Ex vivo studies further suggest that a few hours of exposure to vasoconstrictor agonists induces inward remodeling in the absence of changes in intraluminal pressure. Here we report that a short, 10-min, topical exposure to serotonin (5-HT) + N(ω)-nitro-l-arginine methyl ester hydrochloride (l-NAME) was sufficient to initiate inward remodeling processes in rat cremasteric feed arterioles (100-200 μm lumen diameter), in vivo. Addition of the transglutaminase inhibitor, cystamine, blocked the in vivo remodeling. We further demonstrate that, in isolated arterioles, 5-HT + l-NAME activates transglutaminases and modulates the phosphorylation state of cofilin, a regulator of actin depolymerization. The 5-HT + l-NAME-induced remodeling process in isolated arterioles was also inhibited by an inhibitor of Lim Kinase, the kinase that phosphorylates and inactivates cofilin. Therefore, our results indicate that a brief vasoconstriction induced by 5-HT + l-NAME is able to reduce the passive structural diameter of arterioles through processes that are dependent on the activation of transglutaminases and Lim kinase, and the subsequent phosphorylation of cofilin.
Collapse
Affiliation(s)
- Christopher A Foote
- Dalton Cardiovascular Research Center, and Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri
| | - Jorge A Castorena-Gonzalez
- Dalton Cardiovascular Research Center, and Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri; Department of Biological Engineering, University of Missouri-Columbia, Columbia, Missouri; and
| | - Marius C Staiculescu
- Dalton Cardiovascular Research Center, and Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri
| | - Philip S Clifford
- College of Applied Health Sciences, University of Illinois at Chicago, Chicago, Illinois
| | - Michael A Hill
- Dalton Cardiovascular Research Center, and Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri
| | - Gerald A Meininger
- Dalton Cardiovascular Research Center, and Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri
| | - Luis A Martinez-Lemus
- Dalton Cardiovascular Research Center, and Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri; Department of Biological Engineering, University of Missouri-Columbia, Columbia, Missouri; and
| |
Collapse
|
47
|
Haltia O, Törmänen S, Eräranta A, Jokihaara J, Nordhausen K, Rysä J, Ruskoaho H, Tikkanen I, Mustonen J, Pörsti I. Vasopeptidase Inhibition Corrects the Structure and Function of the Small Arteries in Experimental Renal Insufficiency. J Vasc Res 2015; 52:94-102. [PMID: 26184548 DOI: 10.1159/000431368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 05/05/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND We studied whether vasopeptidase inhibition corrects the structure and function of the small arteries in experimental chronic renal insufficiency (CRI). METHODS After 5/6 nephrectomy (NX) surgery was performed on rats, there was a 14-week follow-up, allowing CRI to become established. Omapatrilat (40 mg/kg/day in chow) was then given for 8 weeks, and the small mesenteric arterial rings were investigated in vitro using wire and pressure myographs. RESULTS Plasma and ventricular B-type natriuretic peptide (BNP) concentrations were increased 2- to 2.7-fold, while systolic blood pressure (BP) increased by 32 mm Hg after NX. Omapatrilat treatment normalized the BNP and reduced the BP by 45 mm Hg in the NX rats. Endothelium-dependent vasorelaxation was impaired but the response to acetylcholine was normalized after omapatrilat treatment. Vasorelaxations induced by nitroprusside, isoprenaline and levcromakalim were enhanced after omapatrilat, and the responses were even more pronounced than in untreated sham-operated rats. Arterial wall thickness and wall-to-lumen ratio were increased after NX, whereas omapatrilat normalized these structural features and improved the strain-stress relationship in the small arteries; this suggests improved arterial elastic properties. CONCLUSION Omapatrilat treatment reduced BP, normalized volume overload, improved vasorelaxation and corrected the dimensions and passive elastic properties of the small arteries in the NX rats. Therefore, we consider vasopeptidase inhibition to be an effective treatment for CRI-induced changes in the small arteries.
Collapse
Affiliation(s)
- Olli Haltia
- Schools of Medicine, University of Tampere, Tampere, Finland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Bender SB, Castorena-Gonzalez JA, Garro M, Reyes-Aldasoro CC, Sowers JR, DeMarco VG, Martinez-Lemus LA. Regional variation in arterial stiffening and dysfunction in Western diet-induced obesity. Am J Physiol Heart Circ Physiol 2015; 309:H574-82. [PMID: 26092984 DOI: 10.1152/ajpheart.00155.2015] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 06/18/2015] [Indexed: 12/11/2022]
Abstract
Increased central vascular stiffening, assessed in vivo by determination of pulse wave velocity (PWV), is an independent predictor of cardiovascular event risk. Recent evidence demonstrates that accelerated aortic stiffening occurs in obesity; however, little is known regarding stiffening of other disease-relevant arteries or whether regional variation in arterial stiffening occurs in this setting. We addressed this gap in knowledge by assessing femoral PWV in vivo in conjunction with ex vivo analyses of femoral and coronary structure and function in a mouse model of Western diet (WD; high-fat/high-sugar)-induced obesity and insulin resistance. WD feeding resulted in increased femoral PWV in vivo. Ex vivo analysis of femoral arteries revealed a leftward shift in the strain-stress relationship, increased modulus of elasticity, and decreased compliance indicative of increased stiffness following WD feeding. Confocal and multiphoton fluorescence microscopy revealed increased femoral stiffness involving decreased elastin/collagen ratio in conjunction with increased femoral transforming growth factor-β (TGF-β) content in WD-fed mice. Further analysis of the femoral internal elastic lamina (IEL) revealed a significant reduction in the number and size of fenestrae with WD feeding. Coronary artery stiffness and structure was unchanged by WD feeding. Functionally, femoral, but not coronary, arteries exhibited endothelial dysfunction, whereas coronary arteries exhibited increased vasoconstrictor responsiveness not present in femoral arteries. Taken together, our data highlight important regional variations in the development of arterial stiffness and dysfunction associated with WD feeding. Furthermore, our results suggest TGF-β signaling and IEL fenestrae remodeling as potential contributors to femoral artery stiffening in obesity.
Collapse
Affiliation(s)
- Shawn B Bender
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri; Department of Biomedical Sciences, University of Missouri School of Medicine, Columbia, Missouri; Dalton Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, Missouri
| | - Jorge A Castorena-Gonzalez
- Dalton Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, Missouri; Department of Biological Engineering, University of Missouri, Columbia, Missouri
| | - Mona Garro
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri; Department of Medicine-Endocrinology, Diabetes and Metabolism University of Missouri School of Medicine, Columbia, Missouri
| | | | - James R Sowers
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri; Dalton Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, Missouri; Department of Medicine-Endocrinology, Diabetes and Metabolism University of Missouri School of Medicine, Columbia, Missouri, Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri; and
| | - Vincent G DeMarco
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri; Department of Medicine-Endocrinology, Diabetes and Metabolism University of Missouri School of Medicine, Columbia, Missouri, Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri; and
| | - Luis A Martinez-Lemus
- Dalton Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, Missouri; Department of Biological Engineering, University of Missouri, Columbia, Missouri; Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri; and
| |
Collapse
|
49
|
Vasodilatory effect of a novel Rho-kinase inhibitor, DL0805-2, on the rat mesenteric artery and its potential mechanisms. Cardiovasc Drugs Ther 2015; 28:415-24. [PMID: 25086815 DOI: 10.1007/s10557-014-6544-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE In the present study, we investigated the vasodilatory effect of a novel scaffold Rho-kinase inhibitor, DL0805-2, on isolated rat arterial rings including mesenteric, ventral tail, and renal arteries. We also examined the potential mechanisms of its vasodilatory action using mesenteric artery rings. METHODS A DMT multiwire myograph system was used to test the tension of isolated small arteries. Several drugs were employed to verify the underlying mechanisms. RESULTS DL0805-2 (10(-7)-10(-4) M) inhibited KCl (60 mM)-induced vasoconstriction in three types of small artery rings (pEC50: 5.84 ± 0.03, 5.39 ± 0.03, and 5.67 ± 0.02 for mesenteric, renal, and ventral tail artery rings, respectively). Pre-incubation with DL0805-2 (1, 3, or 10 μM) attenuated KCl (10-60 mM) and angiotensin II (AngII; 10(-6) M)-induced vasoconstriction in mesenteric artery rings. The relaxant effect on the rat mesenteric artery was partially endothelium-dependent (pEC50: 6.02 ± 0.05 for endothelium-intact and 5.72 ± 0.06 for endothelium-denuded). The influx and release of Ca(2+) were inhibited by DL0805-2. In addition, the increased phosphorylation levels of myosin light chain (MLC) and myosin-binding subunit of myosin phosphatase (MYPT1) induced by AngII were blocked by DL0805-2. However, DL0805-2 had little effect on K(+) channels. CONCLUSIONS The present results demonstrate that DL0805-2 has a vasorelaxant effect on isolated rat small arteries and may exert its action through the endothelium, Ca(2+) channels, and the Rho/ROCK pathway.
Collapse
|
50
|
Turczyńska KM, Swärd K, Hien TT, Wohlfahrt J, Mattisson IY, Ekman M, Nilsson J, Sjögren J, Murugesan V, Hultgårdh-Nilsson A, Cidad P, Hellstrand P, Pérez-García MT, Albinsson S. Regulation of Smooth Muscle Dystrophin and Synaptopodin 2 Expression by Actin Polymerization and Vascular Injury. Arterioscler Thromb Vasc Biol 2015; 35:1489-97. [DOI: 10.1161/atvbaha.114.305065] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 03/27/2015] [Indexed: 01/25/2023]
Abstract
Objective—
Actin dynamics in vascular smooth muscle is known to regulate contractile differentiation and may play a role in the pathogenesis of vascular disease. However, the list of genes regulated by actin polymerization in smooth muscle remains incomprehensive. Thus, the objective of this study was to identify actin-regulated genes in smooth muscle and to demonstrate the role of these genes in the regulation of vascular smooth muscle phenotype.
Approach and Results—
Mouse aortic smooth muscle cells were treated with an actin-stabilizing agent, jasplakinolide, and analyzed by microarrays. Several transcripts were upregulated including both known and previously unknown actin-regulated genes. Dystrophin and synaptopodin 2 were selected for further analysis in models of phenotypic modulation and vascular disease. These genes were highly expressed in differentiated versus synthetic smooth muscle and their expression was promoted by the transcription factors myocardin and myocardin-related transcription factor A. Furthermore, the expression of both synaptopodin 2 and dystrophin was significantly reduced in balloon-injured human arteries. Finally, using a dystrophin mutant
mdx
mouse and synaptopodin 2 knockdown, we demonstrate that these genes are involved in the regulation of smooth muscle differentiation and function.
Conclusions—
This study demonstrates novel genes that are promoted by actin polymerization, that regulate smooth muscle function, and that are deregulated in models of vascular disease. Thus, targeting actin polymerization or the genes controlled in this manner can lead to novel therapeutic options against vascular pathologies that involve phenotypic modulation of smooth muscle cells.
Collapse
Affiliation(s)
- Karolina M. Turczyńska
- From the Department of Experimental Medical Science (K.M.T., K.S., T.T.H., J.W., I.Y.M., M.E., V.M., A.H.-N., P.H., S.A.) and Department of Clinical Science (J.N., J.S.), Lund University, Lund, Sweden; and Departamento de Bioquímica y Biología Molecular y Fisiología and Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain (P.C., M.T.P.-G.)
| | - Karl Swärd
- From the Department of Experimental Medical Science (K.M.T., K.S., T.T.H., J.W., I.Y.M., M.E., V.M., A.H.-N., P.H., S.A.) and Department of Clinical Science (J.N., J.S.), Lund University, Lund, Sweden; and Departamento de Bioquímica y Biología Molecular y Fisiología and Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain (P.C., M.T.P.-G.)
| | - Tran Thi Hien
- From the Department of Experimental Medical Science (K.M.T., K.S., T.T.H., J.W., I.Y.M., M.E., V.M., A.H.-N., P.H., S.A.) and Department of Clinical Science (J.N., J.S.), Lund University, Lund, Sweden; and Departamento de Bioquímica y Biología Molecular y Fisiología and Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain (P.C., M.T.P.-G.)
| | - Johan Wohlfahrt
- From the Department of Experimental Medical Science (K.M.T., K.S., T.T.H., J.W., I.Y.M., M.E., V.M., A.H.-N., P.H., S.A.) and Department of Clinical Science (J.N., J.S.), Lund University, Lund, Sweden; and Departamento de Bioquímica y Biología Molecular y Fisiología and Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain (P.C., M.T.P.-G.)
| | - Ingrid Yao Mattisson
- From the Department of Experimental Medical Science (K.M.T., K.S., T.T.H., J.W., I.Y.M., M.E., V.M., A.H.-N., P.H., S.A.) and Department of Clinical Science (J.N., J.S.), Lund University, Lund, Sweden; and Departamento de Bioquímica y Biología Molecular y Fisiología and Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain (P.C., M.T.P.-G.)
| | - Mari Ekman
- From the Department of Experimental Medical Science (K.M.T., K.S., T.T.H., J.W., I.Y.M., M.E., V.M., A.H.-N., P.H., S.A.) and Department of Clinical Science (J.N., J.S.), Lund University, Lund, Sweden; and Departamento de Bioquímica y Biología Molecular y Fisiología and Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain (P.C., M.T.P.-G.)
| | - Johan Nilsson
- From the Department of Experimental Medical Science (K.M.T., K.S., T.T.H., J.W., I.Y.M., M.E., V.M., A.H.-N., P.H., S.A.) and Department of Clinical Science (J.N., J.S.), Lund University, Lund, Sweden; and Departamento de Bioquímica y Biología Molecular y Fisiología and Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain (P.C., M.T.P.-G.)
| | - Johan Sjögren
- From the Department of Experimental Medical Science (K.M.T., K.S., T.T.H., J.W., I.Y.M., M.E., V.M., A.H.-N., P.H., S.A.) and Department of Clinical Science (J.N., J.S.), Lund University, Lund, Sweden; and Departamento de Bioquímica y Biología Molecular y Fisiología and Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain (P.C., M.T.P.-G.)
| | - Vignesh Murugesan
- From the Department of Experimental Medical Science (K.M.T., K.S., T.T.H., J.W., I.Y.M., M.E., V.M., A.H.-N., P.H., S.A.) and Department of Clinical Science (J.N., J.S.), Lund University, Lund, Sweden; and Departamento de Bioquímica y Biología Molecular y Fisiología and Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain (P.C., M.T.P.-G.)
| | - Anna Hultgårdh-Nilsson
- From the Department of Experimental Medical Science (K.M.T., K.S., T.T.H., J.W., I.Y.M., M.E., V.M., A.H.-N., P.H., S.A.) and Department of Clinical Science (J.N., J.S.), Lund University, Lund, Sweden; and Departamento de Bioquímica y Biología Molecular y Fisiología and Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain (P.C., M.T.P.-G.)
| | - Pilar Cidad
- From the Department of Experimental Medical Science (K.M.T., K.S., T.T.H., J.W., I.Y.M., M.E., V.M., A.H.-N., P.H., S.A.) and Department of Clinical Science (J.N., J.S.), Lund University, Lund, Sweden; and Departamento de Bioquímica y Biología Molecular y Fisiología and Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain (P.C., M.T.P.-G.)
| | - Per Hellstrand
- From the Department of Experimental Medical Science (K.M.T., K.S., T.T.H., J.W., I.Y.M., M.E., V.M., A.H.-N., P.H., S.A.) and Department of Clinical Science (J.N., J.S.), Lund University, Lund, Sweden; and Departamento de Bioquímica y Biología Molecular y Fisiología and Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain (P.C., M.T.P.-G.)
| | - M. Teresa Pérez-García
- From the Department of Experimental Medical Science (K.M.T., K.S., T.T.H., J.W., I.Y.M., M.E., V.M., A.H.-N., P.H., S.A.) and Department of Clinical Science (J.N., J.S.), Lund University, Lund, Sweden; and Departamento de Bioquímica y Biología Molecular y Fisiología and Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain (P.C., M.T.P.-G.)
| | - Sebastian Albinsson
- From the Department of Experimental Medical Science (K.M.T., K.S., T.T.H., J.W., I.Y.M., M.E., V.M., A.H.-N., P.H., S.A.) and Department of Clinical Science (J.N., J.S.), Lund University, Lund, Sweden; and Departamento de Bioquímica y Biología Molecular y Fisiología and Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain (P.C., M.T.P.-G.)
| |
Collapse
|