1
|
Sugawara Y, Mizuno Y, Oku S, Sawada Y, Goto T. Role of protein kinase D1 in vasoconstriction and haemodynamics in rats. Microvasc Res 2024; 152:104627. [PMID: 37963515 DOI: 10.1016/j.mvr.2023.104627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 10/31/2023] [Accepted: 11/06/2023] [Indexed: 11/16/2023]
Abstract
AIMS Protein kinase D (PKD), once considered an effector of protein kinase C (PKC), now plays many pathophysiological roles in various tissues. However, little is known about role of PKD in vascular function. We investigated the role of PKD in contraction of rat aorta and human aortic smooth muscle cells (HASMCs) and in haemodynamics in rats. METHODS AND RESULTS Isometric tension of rat aortic was measured to examine norepinephrine-induced contraction in the presence of PKD, PKC and Rho-kinase inhibitors. Phosphorylation of PKD1, myosin targeting subunit-1 (MYPT1), myosin light chain (MLC), CPI-17 and heat-shock protein 27 (HSP27), and actin polymerization were measured in the aorta. Phosphorylation of MYPT1 and MLC was also measured in HASMCs knocked down with specific siRNAs of PKD 1, 2 and 3. Intracellular calcium concentrations and cell shortening were measured in HASMCs. Norepinephrine-induced aortic contraction was accompanied by increased phosphorylation of PKD1, MYPT1 and MLC and actin polymerization, all of which were attenuated with PKD inhibitor CRT0066101. PKD1 phosphorylation was not inhibited by PKC inhibitor, chelerythrine or Rho kinase inhibitor, fasudil. In HASMCs, the phosphorylation of MYPT1 and MLC was attenuated by PKD1, but not PKD2, 3 knockdown. In HASMCs, CRT0066101 inhibited norepinephrine-induced cell shortening without affecting calcium concentration. Administration of CRT0066101 decreased systemic vascular resistance and blood pressure without affecting cardiac output in rats. CONCLUSIONS PKD1 may play roles in aorta contraction and haemodynamics via phosphorylation of MYPT1 and actin polymerization in a calcium-independent manner.
Collapse
Affiliation(s)
- Yoh Sugawara
- Department of Anaesthesiology and Critical Care Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yusuke Mizuno
- Department of Anaesthesiology and Critical Care Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| | - Shinya Oku
- Department of Anaesthesiology and Critical Care Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yuri Sawada
- Department of Anaesthesiology and Critical Care Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Takahisa Goto
- Department of Anaesthesiology and Critical Care Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
2
|
Coccarelli A, Pant S, Polydoros I, Harraz OF. A new model for evaluating pressure-induced vascular tone in small cerebral arteries. Biomech Model Mechanobiol 2024; 23:271-286. [PMID: 37925376 PMCID: PMC10901969 DOI: 10.1007/s10237-023-01774-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 09/17/2023] [Indexed: 11/06/2023]
Abstract
The capacity of small cerebral arteries (SCAs) to adapt to pressure fluctuations has a fundamental physiological role and appears to be relevant in different pathological conditions. Here, we present a new computational model for quantifying the link, and its contributors, between luminal pressure and vascular tone generation in SCAs. This is assembled by combining a chemical sub-model, representing pressure-induced smooth muscle cell (SMC) signalling, with a mechanical sub-model for the tone generation and its transduction at tissue level. The devised model can accurately reproduce the impact of luminal pressure on different cytoplasmic components involved in myogenic signalling, both in the control case and when combined with some specific pharmacological interventions. Furthermore, the model is also able to capture and predict experimentally recorded pressure-outer diameter relationships obtained for vessels under control conditions, both in a Ca2 + -free bath and under drug inhibition. The modularity of the proposed framework allows the integration of new components for the study of a broad range of processes involved in the vascular function.
Collapse
Affiliation(s)
- Alberto Coccarelli
- Zienkiewicz Institute for Modelling, Data and AI, Faculty of Science and Engineering, Swansea University, Swansea, UK.
| | - Sanjay Pant
- Zienkiewicz Institute for Modelling, Data and AI, Faculty of Science and Engineering, Swansea University, Swansea, UK
| | - Ioannis Polydoros
- Zienkiewicz Institute for Modelling, Data and AI, Faculty of Science and Engineering, Swansea University, Swansea, UK
| | - Osama F Harraz
- Department of Pharmacology, Larner College of Medicine, and Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, USA
| |
Collapse
|
3
|
Mironova GY, Mazumdar N, Hashad AM, El-Lakany MA, Welsh DG. Defining a Role of NADPH Oxidase in Myogenic Tone Development. Microcirculation 2022; 29:e12756. [PMID: 35289024 DOI: 10.1111/micc.12756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 03/07/2022] [Accepted: 03/09/2022] [Indexed: 11/28/2022]
Abstract
OBJECTIVE The myogenic response sets the foundation for blood flow control. Recent findings suggest a role for G-protein coupled receptors (GPCR) and signaling pathways tied to the generation of reactive oxygen species (ROS). In this regard, this study ascertained the impact of NADPH oxidase (Nox) on myogenic tone in rat cerebral resistance arteries. METHODS The study employed real-time qPCR (RT-qPCR), pressure myography, and immunohistochemistry. RESULTS Gq blockade abolished myogenic tone in rat cerebral arteries, linking GPCR to mechanosensation. Subsequent work revealed that general (TEMPOL) and mitochondrial specific (MitoTEMPO) ROS scavengers had little impact on myogenic tone, whereas apocynin, a broad spectrum Nox inhibitor, initiated transient dilation. RT-qPCR revealed Nox1 and Nox2 mRNA expression in smooth muscle cells. Pressure myography defined Nox1 rather than Nox2 is facilitating myogenic tone. We rationalized that Nox1-generated ROS was initiating this response by impairing the ability of the CaV 3.2 channel to elicit negative feedback via BKCa . This hypothesis was confirmed in functional experiments. The proximity ligation assay further revealed that Nox1 and CaV 3.2 colocalize within 40 nm of one another. CONCLUSIONS Our data highlight that vascular pressurization augments Nox1 activity and ensuing ROS production facilitates myogenic tone by limiting Ca2+ influx via CaV 3.2.
Collapse
Affiliation(s)
- Galina Yu Mironova
- Robarts Research Institute and Dept. of Physiology & Pharmacology, The University of Western Ontario, London, Ontario, Canada
| | - Neil Mazumdar
- Robarts Research Institute and Dept. of Physiology & Pharmacology, The University of Western Ontario, London, Ontario, Canada
| | - Ahmed M Hashad
- Robarts Research Institute and Dept. of Physiology & Pharmacology, The University of Western Ontario, London, Ontario, Canada.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Mohammed A El-Lakany
- Robarts Research Institute and Dept. of Physiology & Pharmacology, The University of Western Ontario, London, Ontario, Canada.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Donald G Welsh
- Robarts Research Institute and Dept. of Physiology & Pharmacology, The University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
4
|
Regulation of myosin light-chain phosphorylation and its roles in cardiovascular physiology and pathophysiology. Hypertens Res 2022; 45:40-52. [PMID: 34616031 DOI: 10.1038/s41440-021-00733-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 06/19/2021] [Accepted: 07/08/2021] [Indexed: 01/22/2023]
Abstract
The regulation of muscle contraction is a critical function in the cardiovascular system, and abnormalities may be life-threatening or cause illness. The common basic mechanism in muscle contraction is the interaction between the protein filaments myosin and actin. Although this interaction is primarily regulated by intracellular Ca2+, the primary targets and intracellular signaling pathways differ in vascular smooth muscle and cardiac muscle. Phosphorylation of the myosin regulatory light chain (RLC) is a primary molecular switch for smooth muscle contraction. The equilibrium between phosphorylated and unphosphorylated RLC is dynamically achieved through two enzymes, myosin light chain kinase, a Ca2+-dependent enzyme, and myosin phosphatase, which modifies the Ca2+ sensitivity of contractions. In cardiac muscle, the primary target protein for Ca2+ is troponin C on thin filaments; however, RLC phosphorylation also plays a modulatory role in contraction. This review summarizes recent advances in our understanding of the regulation, physiological function, and pathophysiological involvement of RLC phosphorylation in smooth and cardiac muscles.
Collapse
|
5
|
Dwaib HS, Ajouz G, AlZaim I, Rafeh R, Mroueh A, Mougharbil N, Ragi ME, Refaat M, Obeid O, El-Yazbi AF. Phosphorus Supplementation Mitigates Perivascular Adipose Inflammation-Induced Cardiovascular Consequences in Early Metabolic Impairment. J Am Heart Assoc 2021; 10:e023227. [PMID: 34873915 PMCID: PMC9075232 DOI: 10.1161/jaha.121.023227] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background The complexity of the interaction between metabolic dysfunction and cardiovascular complications has long been recognized to extend beyond simple perturbations of blood glucose levels. Yet, structured interventions targeting the root pathologies are not forthcoming. Growing evidence implicates the inflammatory changes occurring in perivascular adipose tissue (PVAT) as early instigators of cardiovascular deterioration. Methods and Results We used a nonobese prediabetic rat model with localized PVAT inflammation induced by hypercaloric diet feeding, which dilutes inorganic phosphorus (Pi) to energy ratio by 50%, to investigate whether Pi supplementation ameliorates the early metabolic impairment. A 12‐week Pi supplementation at concentrations equivalent to and twice as much as that in the control diet was performed. The localized PVAT inflammation was reversed in a dose‐dependent manner. The increased expression of UCP1 (uncoupling protein1), HIF‐1α (hypoxia inducible factor‐1α), and IL‐1β (interleukin‐1β), representing the hallmark of PVAT inflammation in this rat model, were reversed, with normalization of PVAT macrophage polarization. Pi supplementation restored the metabolic efficiency consistent with its putative role as an UCP1 inhibitor. Alongside, parasympathetic autonomic and cerebrovascular dysfunction function observed in the prediabetic model was reversed, together with the mitigation of multiple molecular and histological cardiovascular damage markers. Significantly, a Pi‐deficient control diet neither induced PVAT inflammation nor cardiovascular dysfunction, whereas Pi reinstatement in the diet after a 10‐week exposure to a hypercaloric low‐Pi diet ameliorated the dysfunction. Conclusions Our present results propose Pi supplementation as a simple intervention to reverse PVAT inflammation and its early cardiovascular consequences, possibly through the interference with hypercaloric‐induced increase in UCP1 expression/activity.
Collapse
Affiliation(s)
- Haneen S Dwaib
- Department of Pharmacology and Toxicology Faculty of Medicine The American University of Beirut Beirut Lebanon.,Department of Nutrition and Food Sciences Faculty of Agriculture and Food Sciences The American University of Beirut Beirut Lebanon
| | - Ghina Ajouz
- Department of Pharmacology and Toxicology Faculty of Medicine The American University of Beirut Beirut Lebanon
| | - Ibrahim AlZaim
- Department of Pharmacology and Toxicology Faculty of Medicine The American University of Beirut Beirut Lebanon.,Department of Biochemistry and Molecular Genetics Faculty of Medicine The American University of Beirut Beirut Lebanon
| | - Rim Rafeh
- Department of Pharmacology and Toxicology Faculty of Medicine The American University of Beirut Beirut Lebanon
| | - Ali Mroueh
- INSERM UMR 1260 Regenerative Nanomedicine FMTSUniversity of Strasbourg Strasbourg France
| | - Nahed Mougharbil
- Department of Pharmacology and Toxicology Faculty of Medicine The American University of Beirut Beirut Lebanon
| | - Marie-Elizabeth Ragi
- Department of Nutrition and Food Sciences Faculty of Agriculture and Food Sciences The American University of Beirut Beirut Lebanon
| | - Marwan Refaat
- Department of Biochemistry and Molecular Genetics Faculty of Medicine The American University of Beirut Beirut Lebanon.,Division of Cardiology Department of Internal Medicine Faculty of Medicine The American University of Beirut Beirut Lebanon
| | - Omar Obeid
- Department of Nutrition and Food Sciences Faculty of Agriculture and Food Sciences The American University of Beirut Beirut Lebanon
| | - Ahmed F El-Yazbi
- Department of Pharmacology and Toxicology Faculty of Medicine The American University of Beirut Beirut Lebanon.,Department of Pharmacology and Toxicology Faculty of Pharmacy Alexandria University Alexandria Egypt.,Faculty of Pharmacy Al-Alamein International University Alamein Egypt
| |
Collapse
|
6
|
EPAC in Vascular Smooth Muscle Cells. Int J Mol Sci 2020; 21:ijms21145160. [PMID: 32708284 PMCID: PMC7404248 DOI: 10.3390/ijms21145160] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/09/2020] [Accepted: 07/19/2020] [Indexed: 02/07/2023] Open
Abstract
Vascular smooth muscle cells (VSMCs) are major components of blood vessels. They regulate physiological functions, such as vascular tone and blood flow. Under pathological conditions, VSMCs undergo a remodeling process known as phenotypic switching. During this process, VSMCs lose their contractility and acquire a synthetic phenotype, where they over-proliferate and migrate from the tunica media to the tunica interna, contributing to the occlusion of blood vessels. Since their discovery as effector proteins of cyclic adenosine 3′,5′-monophosphate (cAMP), exchange proteins activated by cAMP (EPACs) have been shown to play vital roles in a plethora of pathways in different cell systems. While extensive research to identify the role of EPAC in the vasculature has been conducted, much remains to be explored to resolve the reported discordance in EPAC’s effects. In this paper, we review the role of EPAC in VSMCs, namely its regulation of the vascular tone and phenotypic switching, with the likely involvement of reactive oxygen species (ROS) in the interplay between EPAC and its targets/effectors.
Collapse
|
7
|
Bai X, Mangum K, Kakoki M, Smithies O, Mack CP, Taylor JM. GRAF3 serves as a blood volume-sensitive rheostat to control smooth muscle contractility and blood pressure. Small GTPases 2020; 11:194-203. [PMID: 29099324 PMCID: PMC7549679 DOI: 10.1080/21541248.2017.1375602] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Vascular resistance is a major determinant of BP and is controlled, in large part, by RhoA-dependent smooth muscle cell (SMC) contraction within small peripheral arterioles and previous studies from our lab indicate that GRAF3 is a critical regulator of RhoA in vascular SMC. The elevated contractile responses we observed in GRAF3 deficient vessels coupled with the hypertensive phenotype provided a mechanistic link for the hypertensive locus recently identified within the GRAF3 gene. On the basis of our previous findings that the RhoA signaling axis also controls SMC contractile gene expression and that GRAF3 expression was itself controlled by this pathway, we postulated that GRAF3 serves as an important counter-regulator of SMC phenotype. Indeed, our new findings presented herein indicate that GRAF3 expression acts as a pressure-sensitive rheostat to control vessel tone by both reducing calcium sensitivity and restraining expression of the SMC-specific contractile proteins that support this function. Collectively, these studies highlight the potential therapeutic value of GRAF3 in the control of human hypertension.
Collapse
Affiliation(s)
- Xue Bai
- Department of Pathology, University of North Carolina, Chapel Hill, NC, USA
| | - Kevin Mangum
- Department of Pathology, University of North Carolina, Chapel Hill, NC, USA
| | - Masao Kakoki
- Department of Pathology, University of North Carolina, Chapel Hill, NC, USA
| | - Oliver Smithies
- Department of Pathology, University of North Carolina, Chapel Hill, NC, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Christopher P. Mack
- Department of Pathology, University of North Carolina, Chapel Hill, NC, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Joan M. Taylor
- Department of Pathology, University of North Carolina, Chapel Hill, NC, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
8
|
Sancho M, Hald BO, Welsh DG. A stepwise approach to resolving small ionic currents in vascular tissue. Am J Physiol Heart Circ Physiol 2020; 318:H632-H638. [PMID: 32004067 DOI: 10.1152/ajpheart.00628.2019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Arterial membrane potential (Vm) is set by an active interplay among ion channels whose principal function is to set contractility through the gating of voltage-operated Ca2+ channels. To garner an understanding of this electrical parameter, the activity of each channel must be established under near-physiological conditions, a significant challenge given their small magnitude. The inward rectifying K+ (KIR) channel is illustrative of the problem, as its outward "physiological" component is almost undetectable. This study describes a stepwise approach to dissect small ionic currents at physiological Vm using endothelial and smooth muscle cells freshly isolated from rat cerebral arteries. We highlight three critical steps, beginning with the voltage clamping of vascular cells bathed in physiological solutions while maintaining a giga-ohm seal. KIR channels are then inhibited (micromolar Ba2+) so that a difference current can be created, once Ba2+ traces are corrected for the changing seal resistance and subtle instrument drift, pulling the reversal potential rightward. The latter is a new procedure and entails the alignment of whole cell current traces at a voltage where KIR is silent and other channels exhibit limited activity. We subsequently introduced corrected and uncorrected currents into computer models of the arterial wall to show how these subtle adjustments markedly impact the importance of KIR in Vm and arterial tone regulation. We argue that this refined approach can be used on an array of vascular ion channels to build a complete picture of how they dynamically interact to set arterial tone in key organs like the brain.NEW & NOTEWORTHY This work describes a stepwise approach to resolve small ionic currents involved in controlling Vm in resistance arteries. Using this new methodology, we particularly resolved the outward component of the KIR current in native vascular cells, voltage clamped in near-physiological conditions. This novel approach can be applied to any other vascular currents and used to better interpret how vascular ion channels cooperate to control arterial tone.
Collapse
Affiliation(s)
- Maria Sancho
- Robarts Research Institute and the Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Bjorn O Hald
- Department of Neuroscience, Translational Neurobiology, University of Copenhagen, Copenhagen, Denmark
| | - Donald G Welsh
- Robarts Research Institute and the Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
9
|
Elkhatib MAW, Mroueh A, Rafeh RW, Sleiman F, Fouad H, Saad EI, Fouda MA, Elgaddar O, Issa K, Eid AH, Eid AA, Abd-Elrahman KS, El-Yazbi AF. Amelioration of perivascular adipose inflammation reverses vascular dysfunction in a model of nonobese prediabetic metabolic challenge: potential role of antidiabetic drugs. Transl Res 2019; 214:121-143. [PMID: 31408626 DOI: 10.1016/j.trsl.2019.07.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/28/2019] [Accepted: 07/22/2019] [Indexed: 12/12/2022]
Abstract
The onset of vascular impairment precedes that of diagnostic hyperglycemia in diabetic patients suggesting a vascular insult early in the course of metabolic dysfunction without a well-defined mechanism. Mounting evidence implicates adipose inflammation in the pathogenesis of insulin resistance and diabetes. It is not certain whether amelioration of adipose inflammation is sufficient to preclude vascular dysfunction in early stages of metabolic disease. Recent findings suggest that antidiabetic drugs, metformin, and pioglitazone, improve vascular function in prediabetic patients, without an indication if this protective effect is mediated by reduction of adipose inflammation. Here, we used a prediabetic rat model with delayed development of hyperglycemia to study the effect of metformin or pioglitazone on adipose inflammation and vascular function. At the end of the metabolic challenge, these rats were neither obese, hypertensive, nor hyperglycemic. However, they showed increased pressor responses to phenylephrine and augmented aortic and mesenteric contraction. Vascular tissues from prediabetic rats showed increased Rho-associated kinase activity causing enhanced calcium sensitization. An elevated level of reactive oxygen species was seen in aortic tissues together with increased Transforming growth factor β1 and Interleukin-1β expression. Although, no signs of systemic inflammation were detected, perivascular adipose inflammation was observed. Adipocyte hypertrophy, increased macrophage infiltration, and elevated Transforming growth factor β1 and Interleukin-1β mRNA levels were seen. Two-week treatment with metformin or pioglitazone or switching to normal chow ameliorated adipose inflammation and vascular dysfunction. Localized perivascular adipose inflammation is sufficient to trigger vascular dysfunction early in the course of diabetes. Interfering with this inflammatory process reverses this early abnormality.
Collapse
Affiliation(s)
- Mohammed A W Elkhatib
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Ali Mroueh
- Department of Pharmacology and Toxicology, Faculty of Medicine, The American University of Beirut, Beirut, Lebanon
| | - Rim W Rafeh
- Department of Pharmacology and Toxicology, Faculty of Medicine, The American University of Beirut, Beirut, Lebanon
| | - Fatima Sleiman
- Department of Pharmacology and Toxicology, Faculty of Medicine, The American University of Beirut, Beirut, Lebanon
| | - Hosny Fouad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Evan I Saad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Mohamed A Fouda
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Ola Elgaddar
- Department of Chemical Pathology, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Khodr Issa
- Department of Pharmacology and Toxicology, Faculty of Medicine, The American University of Beirut, Beirut, Lebanon
| | - Ali H Eid
- Department of Pharmacology and Toxicology, Faculty of Medicine, The American University of Beirut, Beirut, Lebanon; Department of Biomedical Sciences, Qatar University, Doha, Qatar
| | - Assaad A Eid
- Department of Anatomy, Cell Biology and Physiology, Faculty of Medicine, The American University of Beirut, Beirut, Lebanon
| | - Khaled S Abd-Elrahman
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa Brain and Mind Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Ahmed F El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt; Department of Pharmacology and Toxicology, Faculty of Medicine, The American University of Beirut, Beirut, Lebanon.
| |
Collapse
|
10
|
AlFadly ED, Elzahhar PA, Tramarin A, Elkazaz S, Shaltout H, Abu-Serie MM, Janockova J, Soukup O, Ghareeb DA, El-Yazbi AF, Rafeh RW, Bakkar NMZ, Kobeissy F, Iriepa I, Moraleda I, Saudi MN, Bartolini M, Belal AS. Tackling neuroinflammation and cholinergic deficit in Alzheimer's disease: Multi-target inhibitors of cholinesterases, cyclooxygenase-2 and 15-lipoxygenase. Eur J Med Chem 2019; 167:161-186. [DOI: 10.1016/j.ejmech.2019.02.012] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/04/2019] [Accepted: 02/04/2019] [Indexed: 12/31/2022]
|
11
|
Pabbidi MR, Kuppusamy M, Didion SP, Sanapureddy P, Reed JT, Sontakke SP. Sex differences in the vascular function and related mechanisms: role of 17β-estradiol. Am J Physiol Heart Circ Physiol 2018; 315:H1499-H1518. [DOI: 10.1152/ajpheart.00194.2018] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The incidence of cardiovascular disease (CVD) is lower in premenopausal women but increases with age and menopause compared with similarly aged men. Based on the prevalence of CVD in postmenopausal women, sex hormone-dependent mechanisms have been postulated to be the primary factors responsible for the protection from CVD in premenopausal women. Recent Women’s Health Initiative studies, Cochrane Review studies, the Early Versus Late Intervention Trial with Estradiol Study, and the Kronos Early Estrogen Prevention Study have suggested that beneficial effects of hormone replacement therapy (HRT) are seen in women of <60 yr of age and if initiated within <10 yr of menopause. In contrast, the beneficial effects of HRT are not seen in women of >60 yr of age and if commenced after 10 yr of menopause. The higher incidence of CVD and the failure of HRT in postmenopausal aged women could be partly associated with fundamental differences in the vascular structure and function between men and women and in between pre- and postmenopausal women, respectively. In this regard, previous studies from human and animal studies have identified several sex differences in vascular function and associated mechanisms. The female sex hormone 17β-estradiol regulates the majority of these mechanisms. In this review, we summarize the sex differences in vascular structure, myogenic properties, endothelium-dependent and -independent mechanisms, and the role of 17β-estradiol in the regulation of vascular function.
Collapse
Affiliation(s)
- Mallikarjuna R. Pabbidi
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Maniselvan Kuppusamy
- Division of Endocrinology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Sean P. Didion
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Padmaja Sanapureddy
- Department of Primary Care and Medicine, G. V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, Mississippi
| | - Joey T. Reed
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Sumit P. Sontakke
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
12
|
Lubomirov LT, Papadopoulos S, Filipova D, Baransi S, Todorović D, Lake P, Metzler D, Hilsdorf S, Schubert R, Schroeter MM, Pfitzer G. The involvement of phosphorylation of myosin phosphatase targeting subunit 1 (MYPT1) and MYPT1 isoform expression in NO/cGMP mediated differential vasoregulation of cerebral arteries compared to systemic arteries. Acta Physiol (Oxf) 2018; 224:e13079. [PMID: 29694711 DOI: 10.1111/apha.13079] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 03/30/2018] [Accepted: 04/17/2018] [Indexed: 12/23/2022]
Abstract
AIM Constitutive release of NO blunts intrinsic and stimulated contractile activity in cerebral arteries (CA). Here, we explored whether phosphorylation and expression levels of the PKG-sensitive, leucine zipper positive (LZ+ ) splice variants of the regulatory subunit of myosin phosphatase (MYPT1) are involved and whether its expression is associated with higher cGMP sensitivity. METHODS Vascular contractility was investigated by wire myography. Phosphorylation of MYPT1 was determined by Western blotting. RESULTS Constitutive phosphorylation of MYPT1-T696 and T853 was lower and that of S695 and S668 was higher in cerebral arteries from the circulus arteriosus (CA-w) than in femoral arteries (FA), while total MYPT1 expression was not different. In CA-w but not in FA, L-NAME lowered phosphorylation of S695/S668 and increased phosphorylation of T696/T853 and of MLC20 -S19, plus basal tone. The increase in basal tone was attenuated in CA-w and basilar arteries (BA) from heterozygous MYPT1-T696A/+ mice. Compared to FA, expression of the LZ+ -isoform was ~2-fold higher in CA-w coincident with a higher sensitivity to DEA-NONOate, cinaciguat and Y27632 in BA and 8-Br-cGMP (1 μmol/L) in pre-constricted (pCa 6.1) α-toxin permeabilized CAs. In contrast, 6-Bnz-cAMP (10 μmol/L) relaxed BA and FA similarly by ~80%. CONCLUSION Our results indicate that (i) regulation of the intrinsic contractile activity in CA involves phosphorylation of MYPT1 at T696 and S695/S668, (ii) the higher NO/cGMP/PKG sensitivity of CAs can be ascribed to the higher expression level of the LZ+ -MYPT1 isoform and (iii) relaxation by cAMP/PKA pathway is less dependent on the expression level of the LZ+ splice variants of MYPT1.
Collapse
Affiliation(s)
- L. T. Lubomirov
- Institute of Vegetative Physiology; University of Cologne; Cologne Germany
| | - S. Papadopoulos
- Institute of Vegetative Physiology; University of Cologne; Cologne Germany
| | - D. Filipova
- Institute of Vegetative Physiology; University of Cologne; Cologne Germany
| | - S. Baransi
- Institute of Vegetative Physiology; University of Cologne; Cologne Germany
| | - D. Todorović
- Institute of Vegetative Physiology; University of Cologne; Cologne Germany
| | - P. Lake
- Institute of Vegetative Physiology; University of Cologne; Cologne Germany
| | - D. Metzler
- Institute of Vegetative Physiology; University of Cologne; Cologne Germany
| | - S. Hilsdorf
- Institute of Vegetative Physiology; University of Cologne; Cologne Germany
| | - R. Schubert
- Research Division Cardiovascular Physiology; Centre for Biomedicine and Medical Technology Mannheim (CBTM); Ruprecht-Karls-University Heidelberg; Heidelberg Germany
| | - M. M. Schroeter
- Institute of Vegetative Physiology; University of Cologne; Cologne Germany
| | - G. Pfitzer
- Institute of Vegetative Physiology; University of Cologne; Cologne Germany
| |
Collapse
|
13
|
Cardiac Autonomic Neuropathy as a Result of Mild Hypercaloric Challenge in Absence of Signs of Diabetes: Modulation by Antidiabetic Drugs. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:9389784. [PMID: 29643979 PMCID: PMC5831709 DOI: 10.1155/2018/9389784] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 11/21/2017] [Accepted: 11/29/2017] [Indexed: 12/13/2022]
Abstract
Cardiac autonomic neuropathy (CAN) is an early cardiovascular complication of diabetes occurring before metabolic derangement is evident. The cause of CAN remains elusive and cannot be directly linked to hyperglycemia. Recent clinical data report cardioprotective effects of some antidiabetic drugs independent of their hypoglycemic action. Here, we used a rat model receiving limited daily increase in calories from fat (HC diet) to assess whether mild metabolic challenge led to CAN in absence of interfering effects of hyperglycemia, glucose intolerance, or obesity. Rats receiving HC diet for 12 weeks showed reduction in baroreceptor sensitivity and heart rate variability despite lack of change in baseline hemodynamic and cardiovascular structural parameters. Impairment of cardiac autonomic control was accompanied with perivascular adipose inflammation observed as an increased inflammatory cytokine expression, together with increased cardiac oxidative stress, and signaling derangement characteristic of diabetic cardiomyopathy. Two-week treatment with metformin or pioglitazone rectified the autonomic derangement and corrected the molecular changes. Switching rats to normal chow but not to isocaloric amounts of HC for two weeks reversed CAN. As such, we conclude that adipose inflammation due to increased fat intake might underlie development of CAN and, hence, the beneficial effects of metformin and pioglitazone.
Collapse
|
14
|
Lidington D, Kroetsch JT, Bolz SS. Cerebral artery myogenic reactivity: The next frontier in developing effective interventions for subarachnoid hemorrhage. J Cereb Blood Flow Metab 2018; 38:17-37. [PMID: 29135346 PMCID: PMC5757446 DOI: 10.1177/0271678x17742548] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Aneurysmal subarachnoid hemorrhage (SAH) is a devastating cerebral event that kills or debilitates the majority of those afflicted. The blood that spills into the subarachnoid space stimulates profound cerebral artery vasoconstriction and consequently, cerebral ischemia. Thus, once the initial bleeding in SAH is appropriately managed, the clinical focus shifts to maintaining/improving cerebral perfusion. However, current therapeutic interventions largely fail to improve clinical outcome, because they do not effectively restore normal cerebral artery function. This review discusses emerging evidence that perturbed cerebrovascular "myogenic reactivity," a crucial microvascular process that potently dictates cerebral perfusion, is the critical element underlying cerebral ischemia in SAH. In fact, the myogenic mechanism could be the reason why many therapeutic interventions, including "Triple H" therapy, fail to deliver benefit to patients. Understanding the molecular basis for myogenic reactivity changes in SAH holds the key to develop more effective therapeutic interventions; indeed, promising recent advancements fuel optimism that vascular dysfunction in SAH can be corrected to improve outcome.
Collapse
Affiliation(s)
- Darcy Lidington
- 1 Department of Physiology, University of Toronto, Toronto, Canada.,2 Toronto Centre for Microvascular Medicine at TBEP, University of Toronto, Toronto, Canada
| | - Jeffrey T Kroetsch
- 1 Department of Physiology, University of Toronto, Toronto, Canada.,2 Toronto Centre for Microvascular Medicine at TBEP, University of Toronto, Toronto, Canada
| | - Steffen-Sebastian Bolz
- 1 Department of Physiology, University of Toronto, Toronto, Canada.,2 Toronto Centre for Microvascular Medicine at TBEP, University of Toronto, Toronto, Canada.,3 Heart & Stroke/Richard Lewar Centre of Excellence for Cardiovascular Research, University of Toronto, Toronto, Canada
| |
Collapse
|
15
|
Takeya K, Kaneko T, Miyazu M, Takai A. Addition of urea and thiourea to electrophoresis sample buffer improves efficiency of protein extraction from TCA/acetone-treated smooth muscle tissues for phos-tag SDS-PAGE. Electrophoresis 2017; 39:326-333. [PMID: 29072784 DOI: 10.1002/elps.201700394] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 10/18/2017] [Indexed: 01/04/2023]
Abstract
Phosphorylation analysis by using phos-tag technique has been reported to be suitable for highly sensitive quantification of smooth muscle myosin regulatory light chain (LC20 ) phosphorylation. However, there is another factor that will affect the sensitivity of phosphorylation analysis, that is, protein extraction. Here, we optimized the conditions for total protein extraction out of trichloroacetic acid (TCA)-fixed tissues. Standard SDS sample buffer extracted less LC20 , actin and myosin phosphatase targeting subunit 1 (MYPT1) from TCA/acetone treated ciliary muscle strips. On the other hand, sample buffer containing urea and thiourea in addition to lithium dodecyl sulfate (LDS) or SDS extracted those proteins more efficiently, and thus increased the detection sensitivity up to 4-5 fold. Phos-tag SDS-PAGE separated dephosphorylated and phosphorylated LC20 s extracted in LDS/urea/thiourea sample buffer to the same extent as those in standard SDS buffer. We have concluded that LDS (or SDS) /urea/thiourea sample buffer is suitable for highly sensitive phosphorylation analysis in smooth muscle, especially when it is treated with TCA/acetone.
Collapse
Affiliation(s)
- Kosuke Takeya
- Department of Physiology, Asahikawa Medical University, Hokkaido, Japan
| | - Toshiyuki Kaneko
- Department of Physiology, Asahikawa Medical University, Hokkaido, Japan
| | - Motoi Miyazu
- Department of Physiology, Asahikawa Medical University, Hokkaido, Japan
| | - Akira Takai
- Department of Physiology, Asahikawa Medical University, Hokkaido, Japan
| |
Collapse
|
16
|
Lin S, Brozovich FV. MYPT1 isoforms expressed in HEK293T cells are differentially phosphorylated after GTPγS treatment. J Smooth Muscle Res 2017; 52:66-77. [PMID: 27725371 PMCID: PMC5321854 DOI: 10.1540/jsmr.52.66] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Agonist stimulation of smooth muscle is known to activate RhoA/Rho kinase signaling, and
Rho kinase phosphorylates the myosin targeting subunit (MYPT1) of myosin light chain (MLC)
phosphatase at Thr696 and Thr853, which inhibits the activity of MLC phosphatase to
produce a Ca2+ independent increase in MLC phosphorylation and force (Ca2+ sensitization).
Alternative mRNA splicing produces four MYPT1 isoforms, which differ by the presence or
absence of a central insert (CI) and leucine zipper (LZ). This study was designed to
determine if Rho kinase differentially phosphorylates MYPT1 isoforms. In HEK293T cells
expressing each of the four MYPT1 isoforms, we could not detect a change in Thr853 MYPT1
phosphorylation following GTPγS treatment. However, there is differential phosphorylation
of MYPT1 isoforms at Thr696; GTPγS treatment increases MYPT1 phosphorylation for the
CI+LZ- and CI-LZ- MYPT1 isoforms, but not the CI+LZ+ or CI-LZ+ MYPT1 isoforms. These data
could suggest that in smooth muscle Rho kinase differentially phosphorylates MYPT1
isoforms.
Collapse
Affiliation(s)
- Simon Lin
- Mayo Medical School, Department of Cardiovascular Disease, Rochester, MN 55905, USA
| | | |
Collapse
|
17
|
Takeya K. Highly sensitive myosin phosphorylation analysis in the renal afferent arteriole. J Smooth Muscle Res 2017; 52:45-55. [PMID: 27375035 PMCID: PMC5137254 DOI: 10.1540/jsmr.52.45] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The regulation of smooth muscle contraction and relaxation involves phosphorylation and
dephosphorylation of regulatory proteins, particularly myosin. To elucidate the regulatory
mechanisms, analyzing the phosphorylation signal transduction is crucial. Although a
pharmacological approach with selective inhibitors is sensitive and a useful technique, it
leads to speculation regarding a signaling pathway but does not provide direct evidence of
changes at a molecular level. We developed a highly sensitive biochemical technique to
analyze phosphorylation by adapting Phos-tag SDS-PAGE. With this technique, we
successfully analyzed myosin light chain (LC20) phosphorylation in tiny renal
afferent arterioles. In the rat afferent arterioles, endothelin-1 (ET-1) induced
diphosphorylation of LC20 at Ser19 and Thr18 as well as monophosphorylation at
Ser19 via ETB receptor activation. Considering that LC20
diphosphorylation can decrease the rate of dephosphorylation and thus relaxation, we
concluded that LC20 diphosphorylation contributes, at least in part, to the
prolonged contraction induced by ET-1 in the renal afferent arteriole.
Collapse
Affiliation(s)
- Kosuke Takeya
- Department of Physiology, Asahikawa Medical University, Hokkaido, Japan
| |
Collapse
|
18
|
El-Yazbi AF, Ibrahim KS, El-Gowelli HM, El-Deeb NM, El-Mas MM. Modulation by NADPH oxidase of the chronic cardiovascular and autonomic interaction between cyclosporine and NSAIDs in female rats. Eur J Pharmacol 2017; 806:96-104. [DOI: 10.1016/j.ejphar.2017.04.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 04/11/2017] [Accepted: 04/13/2017] [Indexed: 12/23/2022]
|
19
|
El-Yazbi AF, Abd-Elrahman KS. ROK and Arteriolar Myogenic Tone Generation: Molecular Evidence in Health and Disease. Front Pharmacol 2017; 8:87. [PMID: 28280468 PMCID: PMC5322222 DOI: 10.3389/fphar.2017.00087] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 02/10/2017] [Indexed: 12/13/2022] Open
Abstract
The myogenic response is an inherent property of resistance arteries that warrants a relatively constant blood flow in response to changes in perfusion pressure and protect delicate organs from vascular insufficiencies and excessive blood flow. This fundamental phenomenon has been extensively studied aiming to elucidate the underlying mechanisms triggering smooth muscle contraction in response to intraluminal pressure elevation, particularly, Rho-associated kinase (ROK)-mediated Ca2+-independent mechanisms. The size of the resistance arteries limits the capacity to examine changes in protein phosphorylation/expression levels associated with ROK signaling. A highly sensitive biochemical detection approach was beneficial in examining the role of ROK in different force generation mechanisms along the course of myogenic constriction. In this mini review, we summarize recent results showing direct evidence for the contribution of ROK in development of myogenic response at the level of mechanotransduction, myosin light chain phosphatase inhibition and dynamic actin cytoskeleton reorganization. We will also present evidence that alterations in ROK signaling could underlie the progressive loss in myogenic response in a rat model of type 2 diabetes.
Collapse
Affiliation(s)
- Ahmed F El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of BeirutBeirut, Lebanon; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria UniversityAlexandria, Egypt
| | - Khaled S Abd-Elrahman
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria UniversityAlexandria, Egypt; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of OttawaOttawa, ON, Canada
| |
Collapse
|
20
|
Abd-Elrahman KS, Colinas O, Walsh EJ, Zhu HL, Campbell CM, Walsh MP, Cole WC. Abnormal myosin phosphatase targeting subunit 1 phosphorylation and actin polymerization contribute to impaired myogenic regulation of cerebral arterial diameter in the type 2 diabetic Goto-Kakizaki rat. J Cereb Blood Flow Metab 2017; 37:227-240. [PMID: 26721393 PMCID: PMC5363741 DOI: 10.1177/0271678x15622463] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 10/26/2015] [Accepted: 11/17/2015] [Indexed: 12/11/2022]
Abstract
The myogenic response of cerebral resistance arterial smooth muscle to intraluminal pressure elevation is a key physiological mechanism regulating blood flow to the brain. Rho-associated kinase plays a critical role in the myogenic response by activating Ca2+ sensitization mechanisms: (i) Rho-associated kinase inhibits myosin light chain phosphatase by phosphorylating its targeting subunit myosin phosphatase targeting subunit 1 (at T855), augmenting 20 kDa myosin regulatory light chain (LC20) phosphorylation and force generation; and (ii) Rho-associated kinase stimulates cytoskeletal actin polymerization, enhancing force transmission to the cell membrane. Here, we tested the hypothesis that abnormal Rho-associated kinase-mediated myosin light chain phosphatase regulation underlies the dysfunctional cerebral myogenic response of the Goto-Kakizaki rat model of type 2 diabetes. Basal levels of myogenic tone, LC20, and MYPT1-T855 phosphorylation were elevated and G-actin content was reduced in arteries of pre-diabetic 8-10 weeks Goto-Kakizaki rats with normal serum insulin and glucose levels. Pressure-dependent myogenic constriction, LC20, and myosin phosphatase targeting subunit 1 phosphorylation and actin polymerization were suppressed in both pre-diabetic Goto-Kakizaki and diabetic (18-20 weeks) Goto-Kakizaki rats, whereas RhoA, ROK2, and MYPT1 expression were unaffected. We conclude that abnormal Rho-associated kinase-mediated Ca2+ sensitization contributes to the dysfunctional cerebral myogenic response in the Goto-Kakizaki model of type 2 diabetes.
Collapse
Affiliation(s)
- Khaled S Abd-Elrahman
- The Smooth Muscle Research Group, Departments of Physiology & Pharmacology, Libin Cardiovascular Institute & Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Olaia Colinas
- The Smooth Muscle Research Group, Departments of Physiology & Pharmacology, Libin Cardiovascular Institute & Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Emma J Walsh
- The Smooth Muscle Research Group, Departments of Physiology & Pharmacology, Libin Cardiovascular Institute & Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Hai-Lei Zhu
- The Smooth Muscle Research Group, Departments of Physiology & Pharmacology, Libin Cardiovascular Institute & Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Christine M Campbell
- The Smooth Muscle Research Group, Departments of Physiology & Pharmacology, Libin Cardiovascular Institute & Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Michael P Walsh
- The Smooth Muscle Research Group, Department of Biochemistry & Molecular Biology, Libin Cardiovascular Institute & Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - William C Cole
- The Smooth Muscle Research Group, Departments of Physiology & Pharmacology, Libin Cardiovascular Institute & Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Alberta, Canada
| |
Collapse
|
21
|
Krishnamoorthy G, Reimann K, Wangemann P. Ryanodine-induced vasoconstriction of the gerbil spiral modiolar artery depends on the Ca 2+ sensitivity but not on Ca 2+ sparks or BK channels. BMC PHYSIOLOGY 2016; 16:6. [PMID: 27806708 PMCID: PMC5093982 DOI: 10.1186/s12899-016-0026-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 10/13/2016] [Indexed: 01/26/2023]
Abstract
Background In many vascular smooth muscle cells (SMCs), ryanodine receptor-mediated Ca2+ sparks activate large-conductance Ca2+-activated K+ (BK) channels leading to lowered SMC [Ca2+]i and vasodilation. Here we investigated whether Ca2+ sparks regulate SMC global [Ca2+]i and diameter in the spiral modiolar artery (SMA) by activating BK channels. Methods SMAs were isolated from adult female gerbils, loaded with the Ca2+-sensitive flourescent dye fluo-4 and pressurized using a concentric double-pipette system. Ca2+ signals and vascular diameter changes were recorded using a laser-scanning confocal imaging system. Effects of various pharmacological agents on Ca2+ signals and vascular diameter were analyzed. Results Ca2+ sparks and waves were observed in pressurized SMAs. Inhibition of Ca2+ sparks with ryanodine increased global Ca2+ and constricted SMA at 40 cmH2O but inhibition of Ca2+ sparks with tetracaine or inhibition of BK channels with iberiotoxin at 40 cmH2O did not produce a similar effect. The ryanodine-induced vasoconstriction observed at 40 cmH2O was abolished at 60 cmH2O, consistent with a greater Ca2+-sensitivity of constriction at 40 cmH2O than at 60 cmH2O. When the Ca2+-sensitivity of the SMA was increased by prior application of 1 nM endothelin-1, ryanodine induced a robust vasoconstriction at 60 cmH2O. Conclusions The results suggest that Ca2+ sparks, while present, do not regulate vascular diameter in the SMA by activating BK channels and that the regulation of vascular diameter in the SMA is determined by the Ca2+-sensitivity of constriction.
Collapse
Affiliation(s)
- Gayathri Krishnamoorthy
- Anatomy & Physiology Department, Cell Physiology Laboratory, Kansas State University, 228 Coles Hall, Manhattan, Kansas, 66506-5802, USA
| | - Katrin Reimann
- Anatomy & Physiology Department, Cell Physiology Laboratory, Kansas State University, 228 Coles Hall, Manhattan, Kansas, 66506-5802, USA.,Department of Otolaryngology-Head and Neck Surgery, Tübingen Hearing Research Centre, and Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Philine Wangemann
- Anatomy & Physiology Department, Cell Physiology Laboratory, Kansas State University, 228 Coles Hall, Manhattan, Kansas, 66506-5802, USA.
| |
Collapse
|
22
|
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: 298] [Impact Index Per Article: 37.3] [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
|
23
|
Sutherland C, MacDonald JA, Walsh MP. Analysis of phosphorylation of the myosin-targeting subunit of myosin light chain phosphatase by Phos-tag SDS-PAGE. Am J Physiol Cell Physiol 2016; 310:C681-91. [PMID: 26864694 DOI: 10.1152/ajpcell.00327.2015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 02/08/2016] [Indexed: 01/28/2023]
Abstract
Phosphorylation of the myosin-targeting subunit 1 of myosin light chain phosphatase (MYPT1) plays an important role in the regulation of smooth muscle contraction, and several sites of phosphorylation by different protein Ser/Thr kinases have been identified. Furthermore, in some instances, phosphorylation at specific sites affects phosphorylation at neighboring sites, with functional consequences. Characterization of the complex phosphorylation of MYPT1 in tissue samples at rest and in response to contractile and relaxant stimuli is, therefore, challenging. We have exploited Phos-tag SDS-PAGE in combination with Western blotting using antibodies to MYPT1, including phosphospecific antibodies, to separate multiple phosphorylated MYPT1 species and quantify MYPT1 phosphorylation stoichiometry using purified, full-length recombinant MYPT1 phosphorylated by Rho-associated coiled-coil kinase (ROCK) and cAMP-dependent protein kinase (PKA). This approach confirmed that phosphorylation of MYPT1 by ROCK occurs at Thr(697)and Thr(855), PKA phosphorylates these two sites and the neighboring Ser(696)and Ser(854), and prior phosphorylation at Thr(697)and Thr(855)by ROCK precludes phosphorylation at Ser(696)and Ser(854)by PKA. Furthermore, phosphorylation at Thr(697)and Thr(855)by ROCK exposes two other sites of phosphorylation by PKA. Treatment of Triton-skinned rat caudal arterial smooth muscle strips with the membrane-impermeant phosphatase inhibitor microcystin or treatment of intact tissue with the membrane-permeant phosphatase inhibitor calyculin A induced slow, sustained contractions that correlated with phosphorylation of MYPT1 at 7 to ≥10 sites. Phos-tag SDS-PAGE thus provides a suitable and convenient method for analysis of the complex, multisite MYPT1 phosphorylation events involved in the regulation of myosin light chain phosphatase activity and smooth muscle contraction.
Collapse
Affiliation(s)
- Cindy Sutherland
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Justin A MacDonald
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Michael P Walsh
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
24
|
Spray S, Rasmussen MNP, Skovsted GF, Warfvinge K, Sheykhzade M, Edvinsson L. Reduced Mechanical Stretch Induces Enhanced Endothelin B Receptor-Mediated Contractility via Activation of Focal Adhesion Kinase and Extracellular Regulated Kinase 1/2 in Cerebral Arteries from Rat. Basic Clin Pharmacol Toxicol 2016; 119:68-77. [PMID: 26781487 DOI: 10.1111/bcpt.12553] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 12/23/2015] [Indexed: 11/30/2022]
Abstract
Cerebral ischaemia results in enhanced endothelin B (ETB ) receptor-mediated contraction and receptor protein expression in the affected cerebrovascular smooth muscle cells (SMC). Organ culture of cerebral arteries is a method to induce similar alterations in ETB receptor expression. We suggest that rapid and sustained reduction in wall tension/stretch is a possible trigger mechanism for this vascular remodelling. Isolated rat middle cerebral artery (MCA) segments were incubated in a wire myograph with or without mechanical stretch, prior to assessment of their contractile response to the selective ETB receptor agonist sarafotoxin 6c. The involvement of extracellular regulated kinase (ERK) 1/2 and focal adhesion kinase (FAK) was studied by their specific inhibitors U0126 and PF-228, respectively. Compared with their stretched counterparts, unstretched MCA segments showed a significantly increased ETB receptor-mediated contractile response after 12 hr of incubation, which was attenuated by either U0126 or PF-228. The functionally increased ETB -mediated contractility could be attributed to two different mechanisms: (i) a difference in ETB receptor localization from primarily endothelial expression to SMC expression and (ii) an increased calcium sensitivity of the SMCs due to an increased expression of the calcium channel transient receptor potential canonical 1. Collectively, our results present a possible mechanism linking lack of vessel wall stretch/tension to changes in ETB receptor-mediated contractility via triggering of an early mechanosensitive signalling pathway involving ERK1/2 and FAK signalling. A mechanism likely to be an initiating factor for the increased ETB receptor-mediated contractility found after cerebral ischaemia.
Collapse
Affiliation(s)
- Stine Spray
- Department of Clinical Experimental Research, Glostrup Research Institute, Rigshospitalet, Glostrup, Denmark
| | - Marianne N P Rasmussen
- Department of Clinical Experimental Research, Glostrup Research Institute, Rigshospitalet, Glostrup, Denmark
| | - Gry F Skovsted
- Department of Clinical Experimental Research, Glostrup Research Institute, Rigshospitalet, Glostrup, Denmark
| | - Karin Warfvinge
- Department of Clinical Experimental Research, Glostrup Research Institute, Rigshospitalet, Glostrup, Denmark.,Department of Clinical Sciences, Division of Experimental Vascular Research, Lund University, Lund, Sweden
| | - Majid Sheykhzade
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lars Edvinsson
- Department of Clinical Experimental Research, Glostrup Research Institute, Rigshospitalet, Glostrup, Denmark.,Department of Clinical Sciences, Division of Experimental Vascular Research, Lund University, Lund, Sweden
| |
Collapse
|
25
|
El-Yazbi AF, Abd-Elrahman KS, Moreno-Dominguez A. PKC-mediated cerebral vasoconstriction: Role of myosin light chain phosphorylation versus actin cytoskeleton reorganization. Biochem Pharmacol 2015; 95:263-78. [DOI: 10.1016/j.bcp.2015.04.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 04/16/2015] [Indexed: 10/23/2022]
|
26
|
Kinoshita E, Kinoshita-Kikuta E, Koike T. The Cutting Edge of Affinity Electrophoresis Technology. Proteomes 2015; 3:42-55. [PMID: 28248262 PMCID: PMC5302491 DOI: 10.3390/proteomes3010042] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 01/26/2015] [Accepted: 03/11/2015] [Indexed: 11/16/2022] Open
Abstract
Affinity electrophoresis is an important technique that is widely used to separate and analyze biomolecules in the fields of biology and medicine. Both quantitative and qualitative information can be gained through affinity electrophoresis. Affinity electrophoresis can be applied through a variety of strategies, such as mobility shift electrophoresis, charge shift electrophoresis or capillary affinity electrophoresis. These strategies are based on changes in the electrophoretic patterns of biological macromolecules that result from interactions or complex-formation processes that induce changes in the size or total charge of the molecules. Nucleic acid fragments can be characterized through their affinity to other molecules, for example transcriptional factor proteins. Hydrophobic membrane proteins can be identified by means of a shift in the mobility induced by a charged detergent. The various strategies have also been used in the estimation of association/disassociation constants. Some of these strategies have similarities to affinity chromatography, in that they use a probe or ligand immobilized on a supported matrix for electrophoresis. Such methods have recently contributed to profiling of major posttranslational modifications of proteins, such as glycosylation or phosphorylation. Here, we describe advances in analytical techniques involving affinity electrophoresis that have appeared during the last five years.
Collapse
Affiliation(s)
- Eiji Kinoshita
- Department of Functional Molecular Science, Institute of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Hiroshima 734-8553, Japan.
| | - Emiko Kinoshita-Kikuta
- Department of Functional Molecular Science, Institute of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Hiroshima 734-8553, Japan.
| | - Tohru Koike
- Department of Functional Molecular Science, Institute of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Hiroshima 734-8553, Japan.
| |
Collapse
|
27
|
Moreno-Domínguez A, El-Yazbi AF, Zhu HL, Colinas O, Zhong XZ, Walsh EJ, Cole DM, Kargacin GJ, Walsh MP, Cole WC. Cytoskeletal reorganization evoked by Rho-associated kinase- and protein kinase C-catalyzed phosphorylation of cofilin and heat shock protein 27, respectively, contributes to myogenic constriction of rat cerebral arteries. J Biol Chem 2014; 289:20939-52. [PMID: 24914207 PMCID: PMC4110300 DOI: 10.1074/jbc.m114.553743] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 06/03/2014] [Indexed: 12/31/2022] Open
Abstract
Our understanding of the molecular events contributing to myogenic control of diameter in cerebral resistance arteries in response to changes in intravascular pressure, a fundamental mechanism regulating blood flow to the brain, is incomplete. Myosin light chain kinase and phosphatase activities are known to be increased and decreased, respectively, to augment phosphorylation of the 20-kDa regulatory light chain subunits (LC20) of myosin II, which permits cross-bridge cycling and force development. Here, we assessed the contribution of dynamic reorganization of the actin cytoskeleton and thin filament regulation to the myogenic response and serotonin-evoked constriction of pressurized rat middle cerebral arteries. Arterial diameter and the levels of phosphorylated LC(20), calponin, caldesmon, cofilin, and HSP27, as well as G-actin content, were determined. A decline in G-actin content was observed following pressurization from 10 mm Hg to between 40 and 120 mm Hg and in three conditions in which myogenic or agonist-evoked constriction occurred in the absence of a detectable change in LC20 phosphorylation. No changes in thin filament protein phosphorylation were evident. Pressurization reduced G-actin content and elevated the levels of cofilin and HSP27 phosphorylation. Inhibitors of Rho-associated kinase and PKC prevented the decline in G-actin; reduced cofilin and HSP27 phosphoprotein content, respectively; and blocked the myogenic response. Furthermore, phosphorylation modulators of HSP27 and cofilin induced significant changes in arterial diameter and G-actin content of myogenically active arteries. Taken together, our findings suggest that dynamic reorganization of the cytoskeleton involving increased actin polymerization in response to Rho-associated kinase and PKC signaling contributes significantly to force generation in myogenic constriction of cerebral resistance arteries.
Collapse
Affiliation(s)
| | - Ahmed F. El-Yazbi
- From the Smooth Muscle Research Group, Departments of Physiology & Pharmacology and
| | - Hai-Lei Zhu
- From the Smooth Muscle Research Group, Departments of Physiology & Pharmacology and
| | - Olaia Colinas
- From the Smooth Muscle Research Group, Departments of Physiology & Pharmacology and
| | - X. Zoë Zhong
- From the Smooth Muscle Research Group, Departments of Physiology & Pharmacology and
| | - Emma J. Walsh
- From the Smooth Muscle Research Group, Departments of Physiology & Pharmacology and
| | - Dylan M. Cole
- From the Smooth Muscle Research Group, Departments of Physiology & Pharmacology and
| | - Gary J. Kargacin
- From the Smooth Muscle Research Group, Departments of Physiology & Pharmacology and
| | - Michael P. Walsh
- Biochemistry & Molecular Biology, Libin Cardiovascular Institute and Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - William C. Cole
- From the Smooth Muscle Research Group, Departments of Physiology & Pharmacology and
| |
Collapse
|
28
|
Tsai MH, Chang AN, Huang J, He W, Sweeney HL, Zhu M, Kamm KE, Stull JT. Constitutive phosphorylation of myosin phosphatase targeting subunit-1 in smooth muscle. J Physiol 2014; 592:3031-51. [PMID: 24835173 DOI: 10.1113/jphysiol.2014.273011] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Smooth muscle contraction initiated by myosin regulatory light chain (RLC) phosphorylation is dependent on the relative activities of Ca(2+)-calmodulin-dependent myosin light chain kinase (MLCK) and myosin light chain phosphatase (MLCP). We have investigated the physiological role of the MLCP regulatory subunit MYPT1 in bladder smooth muscle containing a smooth muscle-specific deletion of MYPT1 in adult mice. Deep-sequencing analyses of mRNA and immunoblotting revealed that MYPT1 depletion reduced the amount of PP1cδ with no compensatory changes in expression of other MYPT1 family members. Phosphatase activity towards phosphorylated smooth muscle heavy meromyosin was proportional to the amount of PP1cδ in total homogenates from wild-type or MYPT1-deficient tissues. Isolated MYPT1-deficient tissues from MYPT1(SM-/-) mice contracted with moderate differences in response to KCl and carbachol treatments, and relaxed rapidly with comparable rates after carbachol removal and only 1.5-fold slower after KCl removal. Measurements of phosphorylated proteins in the RLC signalling and actin polymerization modules during contractions revealed moderate changes. Using a novel procedure to quantify total phosphorylation of MYPT1 at Thr696 and Thr853, we found substantial phosphorylation in wild-type tissues under resting conditions, predicting attenuation of MLCP activity. Reduced PP1cδ activity in MYPT1-deficient tissues may be similar to the attenuated MLCP activity in wild-type tissues resulting from constitutively phosphorylated MYPT1. Constitutive phosphorylation of MYPT1 Thr696 and Thr853 may thus represent a physiological mechanism acting in concert with agonist-induced MYPT1 phosphorylation to inhibit MLCP activity. In summary, MYPT1 deficiency may not cause significant derangement of smooth muscle contractility because the effective MLCP activity is not changed.
Collapse
Affiliation(s)
- Ming-Ho Tsai
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Audrey N Chang
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jian Huang
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Weiqi He
- Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing, China
| | - H Lee Sweeney
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Minsheng Zhu
- Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing, China
| | - Kristine E Kamm
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - James T Stull
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| |
Collapse
|
29
|
Khasnis M, Nakatomi A, Gumpper K, Eto M. Reconstituted human myosin light chain phosphatase reveals distinct roles of two inhibitory phosphorylation sites of the regulatory subunit, MYPT1. Biochemistry 2014; 53:2701-9. [PMID: 24712327 PMCID: PMC4010256 DOI: 10.1021/bi5001728] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
![]()
The myosin light chain phosphatase
(MLCP) is a cytoskeleton-associated
protein phosphatase-1 (PP1) holoenzyme and a RhoA/ROCK effector, regulating
cytoskeletal reorganization. ROCK-induced phosphorylation of the MLCP
regulatory subunit (MYPT1) at two sites, Thr696 and Thr853, suppresses
the activity, although little is known about the difference in the
role. Here, we developed a new method for the preparation of the recombinant
human MLCP complex and determined the molecular and cellular basis
of inhibitory phosphorylation. The recombinant MLCP partially purified
from mammalian cell lysates retained characteristics of the native
enzyme, such that it was fully active without Mn2+ and
sensitive to PP1 inhibitor compounds. Selective thio-phosphorylation
of MYPT1 at Thr696 with ROCK inhibited the MLCP activity 30%, whereas
the Thr853 thio-phosphorylation did not alter the phosphatase activity.
Interference with the docking of phospho-Thr696 at the active site
weakened the inhibition, suggesting selective autoinhibition induced
by phospho-Thr696. Both Thr696 and Thr853 sites underwent autodephosphorylation.
Compared with that of Thr853, phosphorylation of Thr696 was more stable,
and it facilitated Thr853 phosphorylation. Endogenous MYPT1 at Thr696
was spontaneously phosphorylated in quiescent human leiomyosarcoma
cells. Serum stimulation of the cells resulted in dissociation of
MYPT1 from myosin and PP1C in parallel with an increase in the level
of Thr853 phosphorylation. The C-terminal domain of human MYPT1(495–1030)
was responsible for the binding to the N-terminal portion of myosin
light meromyosin. The spontaneous phosphorylation at Thr696 may adjust
the basal activity of cellular MLCP and affect the temporal phosphorylation
at Thr853 that is synchronized with myosin targeting.
Collapse
Affiliation(s)
- Mukta Khasnis
- Department of Molecular Physiology and Biophysics, Thomas Jefferson University Jefferson Medical School , and Kimmel Cancer Center , 1020 Locust Street, Philadelphia, Pennsylvania 19107, United States
| | | | | | | |
Collapse
|
30
|
Suppression of the Rho/Rho-kinase pathway and prevention of cerebral vasospasm by combination treatment with statin and fasudil after subarachnoid hemorrhage in rabbit. Transl Stroke Res 2013; 4:368-74. [PMID: 23658597 PMCID: PMC3644406 DOI: 10.1007/s12975-012-0247-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 12/25/2012] [Indexed: 12/19/2022]
Abstract
The Rho/Rho-kinase pathway is considered important in the pathogenesis of sustained smooth muscle cell contraction during cerebral vasospasm after aneurysmal subarachnoid hemorrhage (SAH). The aims of this study were to investigate whether combination treatment, with pitavastatin as an inhibitor of RhoA and fasudil as an inhibitor of Rho-kinase, prevents the cerebral vasospasm. SAH was simulated using the double-hemorrhage rabbit model, and pitavastatin, or fasudil, or both (combination treatment) were administrated. The basilar artery (BA) cross-sectional area only in the combination treatment group was statistically larger than in the SAH group (p < 0.05). BA Rho-kinase, as measured by ELISA, was statistically reduced only in the combination treatment group compared with the SAH group (p < 0.05). In the other two treatment groups, pitavastatin or fasudil treatment group showed larger BA cross-sectional areas and lower value for BA Rho-kinase, but there were no statistically significant differences compared with the SAH group. The expression of endothelial nitric oxide synthase (eNOS), evaluated by immunohistochemistry in the pitavastatin group and the combination group, was higher than in the SAH group. Results indicate that combination treatment could extensively prevent cerebral vasospasm due to the synergic effect of combining pitavastatin and fasudil on the Rho/Rho-kinase pathway and on eNOS.
Collapse
|
31
|
Abstract
The myogenic response has a critical role in regulation of blood flow to the brain. Increased intraluminal pressure elicits vasoconstriction, whereas decreased intraluminal pressure induces vasodilatation, thereby maintaining flow constant over the normal physiologic blood pressure range. Improved understanding of the molecular mechanisms underlying the myogenic response is crucial to identify deficiencies with pathologic consequences, such as cerebral vasospasm, hypertension, and stroke, and to identify potential therapeutic targets. Three mechanisms have been suggested to be involved in the myogenic response: (1) membrane depolarization, which induces Ca(2+) entry, activation of myosin light chain kinase, phosphorylation of the myosin regulatory light chains (LC(20)), increased actomyosin MgATPase activity, cross-bridge cycling, and vasoconstriction; (2) activation of the RhoA/Rho-associated kinase (ROCK) pathway, leading to inhibition of myosin light chain phosphatase by phosphorylation of MYPT1, the myosin targeting regulatory subunit of the phosphatase, and increased LC(20) phosphorylation; and (3) activation of the ROCK and protein kinase C pathways, leading to actin polymerization and the formation of enhanced connections between the actin cytoskeleton, plasma membrane, and extracellular matrix to augment force transmission. This review describes these three mechanisms, emphasizing recent developments regarding the importance of dynamic actin polymerization in the myogenic response of the cerebral vasculature.
Collapse
|
32
|
Moreno-Domínguez A, Colinas O, El-Yazbi A, Walsh EJ, Hill MA, Walsh MP, Cole WC. Ca2+ sensitization due to myosin light chain phosphatase inhibition and cytoskeletal reorganization in the myogenic response of skeletal muscle resistance arteries. J Physiol 2012; 591:1235-50. [PMID: 23230233 DOI: 10.1113/jphysiol.2012.243576] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Abstract The myogenic response of resistance arteries to intravascular pressure elevation is a fundamental physiological mechanism of crucial importance for blood pressure regulation and organ-specific control of blood flow. The importance of Ca(2+) entry via voltage-gated Ca(2+) channels leading to phosphorylation of the 20 kDa myosin regulatory light chains (LC20) in the myogenic response is well established. Recent studies, however, have suggested a role for Ca(2+) sensitization via activation of the RhoA/Rho-associated kinase (ROK) pathway in the myogenic response. The possibility that enhanced actin polymerization is also involved in myogenic vasoconstriction has been suggested. Here, we have used pressurized resistance arteries from rat gracilis and cremaster skeletal muscles to assess the contribution to myogenic constriction of Ca(2+) sensitization due to: (1) phosphorylation of the myosin targeting subunit of myosin light chain phosphatase (MYPT1) by ROK; (2) phosphorylation of the 17 kDa protein kinase C (PKC)-potentiated protein phosphatase 1 inhibitor protein (CPI-17) by PKC; and (3) dynamic reorganization of the actin cytoskeleton evoked by ROK and PKC. Arterial diameter, MYPT1, CPI-17 and LC20 phosphorylation, and G-actin content were determined at varied intraluminal pressures ± H1152, GF109203X or latrunculin B to suppress ROK, PKC and actin polymerization, respectively. The myogenic response was associated with an increase in MYPT1 and LC20 phosphorylation that was blocked by H1152. No change in phospho-CPI-17 content was detected although the PKC inhibitor, GF109203X, suppressed myogenic constriction. Basal LC20 phosphorylation at 10 mmHg was high at ∼40%, increased to a maximal level of ∼55% at 80 mmHg, and exhibited no additional change on further pressurization to 120 and 140 mmHg. Myogenic constriction at 80 mmHg was associated with a decline in G-actin content by ∼65% that was blocked by inhibition of ROK or PKC. Taken together, our findings indicate that two mechanisms of Ca(2+) sensitization (ROK-mediated phosphorylation of MYPT1-T855 with augmentation of LC20 phosphorylation, and a ROK- and PKC-evoked increase in actin polymerization) contribute to force generation in the myogenic response of skeletal muscle arterioles.
Collapse
Affiliation(s)
- Alejandro Moreno-Domínguez
- The Smooth Muscle Research Group, Department of Physiology and Pharmacology, Libin Cardiovascular Institute and Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, 3330 Hospital Drive N.W., Calgary, Alberta, Canada T2N 4N1
| | | | | | | | | | | | | |
Collapse
|
33
|
MacDonald JA, Moffat LD, Al-Ghabkari A, Sutherland C, Walsh MP. Prostate-apoptosis response-4 phosphorylation in vascular smooth muscle. Arch Biochem Biophys 2012; 535:84-90. [PMID: 23219599 DOI: 10.1016/j.abb.2012.11.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Revised: 11/15/2012] [Accepted: 11/21/2012] [Indexed: 12/12/2022]
Abstract
The protein prostate-apoptosis response (Par)-4 has been implicated in the regulation of smooth muscle contraction, based largely on studies with the A7r5 cell line. A mechanism has been proposed whereby Par-4 binding to MYPT1 (the myosin-targeting subunit of myosin light chain phosphatase, MLCP) blocks access of zipper-interacting protein kinase (ZIPK) to Thr697 and Thr855 of MYPT1, whose phosphorylation is associated with MLCP inhibition. Phosphorylation of Par-4 at Thr155 disrupts its interaction with MYPT1, exposing the sites of phosphorylation in MYPT1 and leading to MLCP inhibition and contraction. We tested this "padlock" hypothesis in a well-characterized vascular smooth muscle system, the rat caudal artery. Par-4 was retained in Triton-skinned tissue, suggesting a tight association with the contractile machinery, and indeed Par-4 co-immunoprecipitated with MYPT1. Treatment of Triton-skinned tissue with the phosphatase inhibitor microcystin (MC) evoked phosphorylation of Par-4 at Thr155, but did not induce its dissociation from the contractile machinery. Furthermore, analysis of the time courses of MC-induced phosphorylation of MYPT1 and Par-4 revealed that MYPT1 phosphorylation at Thr697 or Thr855 preceded Par-4 phosphorylation. Par-4 phosphorylation was inhibited by the non-selective kinase inhibitor staurosporine, but not by inhibitors of ZIPK, Rho-associated kinase or protein kinase C. In addition, Par-4 phosphorylation did not occur upon addition of constitutively-active ZIPK to skinned tissue. We conclude that phosphorylation of Par-4 does not regulate contraction of this vascular smooth muscle tissue by inducing dissociation of Par-4 from MYPT1 to allow phosphorylation of MYPT1 and inhibition of MLCP.
Collapse
Affiliation(s)
- Justin A MacDonald
- Smooth Muscle Research Group and Department of Biochemistry & Molecular Biology, University of Calgary, 3280 Hospital Drive NW, Calgary, AB, Canada T2N 4Z6.
| | | | | | | | | |
Collapse
|
34
|
Han YS, Brozovich FV. Altered reactivity of tertiary mesenteric arteries following acute myocardial ischemia. J Vasc Res 2012; 50:100-8. [PMID: 23172397 DOI: 10.1159/000343015] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Accepted: 08/23/2012] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND It is unknown if cardiac ischemia has any deleterious effect on the contractile properties of nonischemic, peripheral vascular beds. Thus, the objective of the present study was to determine whether acute myocardial ischemia results in peripheral vascular dysfunction. METHODS AND RESULTS This study characterized force maintenance and the sensitivity to acetylcholine (ACh)-mediated smooth muscle (SM) relaxation of tertiary (3rd) mesenteric arteries from Sprague-Dawley rats following 30 min of myocardial ischemia. Both the phosphorylation of nonmuscle (NM) light chain (LC) and SM-LCs as well as the expression of myosin phosphatase targeting subunit 1 (MYPT1) were also determined. Our data demonstrate that acute myocardial ischemia resulted in vascular dysfunction of 3rd mesenteric vessels, characterized by decreases in force maintenance, ACh- and cGMP-mediated SM relaxation, the phosphorylation of NM-LCs and SM-LCs, and MYPT1 expression. Ischemia was also associated with an increase in protein polyubiquitination, suggesting that during ischemia MYPT1 is targeted for degradation or proteolysis. CONCLUSION Acute myocardial ischemia produces peripheral vascular dysfunction; the changes in LC phosphorylation and MYPT1 expression result in a decrease in both tone and the sensitivity to NO-mediated SM relaxation of the peripheral vasculature.
Collapse
Affiliation(s)
- Young Soo Han
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN 55905, USA
| | | |
Collapse
|
35
|
Potts LB, Ren Y, Lu G, Kuo E, Ngo E, Kuo L, Hein TW. Constriction of retinal arterioles to endothelin-1: requisite role of rho kinase independent of protein kinase C and L-type calcium channels. Invest Ophthalmol Vis Sci 2012; 53:2904-12. [PMID: 22427601 DOI: 10.1167/iovs.12-9542] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
PURPOSE Although endothelin-1 (ET-1) is a potent vasoconstrictor peptide implicated in several retinal pathologies, the underlying mechanism of vasoconstriction is understood incompletely. We addressed this issue by assessing the contributions of extracellular calcium (Ca²⁺), L-type voltage-operated calcium channels (L-VOCCs), Rho kinase (ROCK), and protein kinase C (PKC) to ET-1-induced constriction of porcine retinal arterioles, all of which have been implicated commonly in vascular smooth muscle contraction. METHODS Porcine retinal arterioles (~50-100 μm) were isolated for vasomotor study and molecular assessment of ROCK isoforms. RESULTS Isolated arterioles developed stable basal tone at 55 cmH₂O luminal pressure and constricted to ET-1 (0.1 nM) with a 40 ± 6% reduction in resting diameter in 20 minutes. In the absence of extraluminal Ca²⁺, arterioles lost basal tone and failed to constrict to ET-1. Although L-VOCC inhibitor nifedipine reduced basal tone and blocked vasoconstriction to PKC activator PDBu, vasoconstriction to ET-1 was unaffected. The broad-spectrum PKC inhibitor Gö-6983 abolished vasoconstriction to PDBu, but did not alter ET-1-induced vasoconstriction or basal tone. Incubation of arterioles with ROCK inhibitor H-1152 abolished basal tone and vasoconstrictions to ET-1 and PDBu. Both ROCK1 and ROCK2 isoforms were expressed in the retinal arteriolar wall. CONCLUSIONS Extracellular Ca²⁺ entry via L-VOCCs and basal ROCK activity play important roles in the maintenance of basal tones of porcine retinal arterioles. ET-1-induced constriction is mediated by extracellular Ca²⁺ entry independent of L-VOCCs and by ROCK activation without the involvement of PKC. However, direct PKC activation can cause vasoconstriction via L-VOCC and ROCK signaling.
Collapse
Affiliation(s)
- Luke B Potts
- Department of Systems Biology and Translational Medicine, College of Medicine, Texas A&M Health Science Center, Temple, Texas, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Sun Z, Li Z, Meininger GA. Mechanotransduction through fibronectin-integrin focal adhesion in microvascular smooth muscle cells: is calcium essential? Am J Physiol Heart Circ Physiol 2012; 302:H1965-73. [PMID: 22427509 DOI: 10.1152/ajpheart.00598.2011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
It is believed that increased transmural pressure exerts force on vascular smooth muscle cells (VSMCs) and triggers Ca(2+) signaling as an initiating event responsible for the arteriolar myogenic response. However, the mechanisms linking the pressure increase to Ca(2+) signaling are unclear. We have shown previously using atomic force microscopy (AFM) that mechanical force induces a VSMC contractile response when applied to single fibronectin (FN; Sun Z, Martinez-Lemus LA, Hill MA, Meininger GA. Am J Physiol Cell Physiol 295; C268-C278, 2008) focal adhesion sites. This current study seeks to determine whether application of force to single focal adhesions can cause a change in VSMC Ca(2+). Experiments were performed in low passage (p3∼10) as well as in freshly isolated skeletal muscle arteriole VSMCs. AFM-attached microbeads (5 μm) were coated with FN or collagen type I (CN-I) or type IV (CN-IV) and placed on a VSMC for 20 min, resulting in formation of a focal adhesion between the cell and the microbead. In low passage VSMCs, mechanically pulling on the FN-coated beads (800∼3000 pN) did not induce a Ca(2+) increase but did cause a contractile response. In freshly isolated VSMCs, application of an FN or CN-I-coated bead onto the cell surface induced global Ca(2+) increases. However, these Ca(2+) increases were not correlated with the application of AFM pulling force to the bead or with the VSMC contractile responses to FN-coupled pulling. Chelating cytosolic Ca(2+) using BAPTA loading had no negative effect on the focal adhesion-related contractile response in both freshly isolated and low passage VSMCs, while the Rho-kinase inhibitor Y27632 abolished the micromyogenic response in both cases. These observations suggest that, in freshly isolated and cultured VSMCs, application of mechanical force to a focal adhesion does not invoke an acute global Ca(2+) increase. On the other hand, our data support a role for Rho-linked signaling mechanism involved in mechanotransduction leading to focal contraction that is independent of the need for a global increase in VSMC Ca(2+).
Collapse
Affiliation(s)
- Zhe Sun
- Dalton Cardiovascular Research Center and Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO 65211, USA.
| | | | | |
Collapse
|
37
|
Kikkawa Y, Matsuo S, Kameda K, Hirano M, Nakamizo A, Sasaki T, Hirano K. Mechanisms underlying potentiation of endothelin-1-induced myofilament Ca(2+) sensitization after subarachnoid hemorrhage. J Cereb Blood Flow Metab 2012; 32:341-52. [PMID: 21952110 PMCID: PMC3272600 DOI: 10.1038/jcbfm.2011.132] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Increased vascular smooth muscle contractility has an important role in the development of cerebral vasospasm after subarachnoid hemorrhage (SAH). Myofilament Ca(2+) sensitivity is a major determinant of smooth muscle contractility. We investigated changes in the Ca(2+)-sensitizing effect of endothelin-1 (ET-1) and the mechanisms underlying ET-1-induced Ca(2+) sensitization after SAH using a rabbit SAH model. After SAH, the contractile response to ET-1 was enhanced, and the ET(A) receptor expression was upregulated in the basilar artery. In α-toxin-permeabilized preparations, ET-1 induced enhanced and prolonged contraction after SAH, suggesting that ET-1-induced Ca(2+) sensitization is potentiated after SAH. Endothelin-1-induced Ca(2+) sensitization became less sensitive to inhibitors of Rho-associated coiled-coil protein kinase (ROCK) and protein kinase C (PKC) after SAH. The expression of PKCα, ROCK2, PKC-potentiated phosphatase inhibitor of 17 kDa (CPI-17) and myosin phosphatase target subunit 1 (MYPT1) was upregulated, and the level of phosphorylation of CPI-17 and MYPT1 was elevated after SAH. This study demonstrated for the first time that the Ca(2+)-sensitizing effect of ET-1 on myofilaments is potentiated after SAH. The increased expression and activity of PKCα, ROCK2, CPI-17, and MYPT1, as well as the upregulation of ET(A) receptor expression are suggested to underlie the enhanced and prolonged Ca(2+) sensitization induced by ET-1.
Collapse
Affiliation(s)
- Yuichiro Kikkawa
- Division of Molecular Cardiology, Research Institute of Angiocardiology, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka, Japan
| | | | | | | | | | | | | |
Collapse
|
38
|
Walsh MP. Vascular smooth muscle myosin light chain diphosphorylation: mechanism, function, and pathological implications. IUBMB Life 2011; 63:987-1000. [PMID: 21990256 DOI: 10.1002/iub.527] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Revised: 05/21/2011] [Accepted: 05/31/2011] [Indexed: 12/17/2022]
Abstract
Smooth muscle contraction is activated primarily by phosphorylation at S19 of the 20-kDa regulatory light chain subunits of myosin II (LC(20) ) catalyzed by Ca(2+) /calmodulin-dependent myosin light chain kinase. Other kinases, for example, integrin-linked kinase (ILK), Rho-associated kinase (ROCK), and zipper-interacting protein kinase (ZIPK), can phosphorylate T18 in addition to S19, which increases the actin-activated myosin MgATPase activity at subsaturating actin concentrations ∼3-fold. These phosphorylatable residues and the amino acid sequence surrounding them are highly conserved throughout the animal kingdom; they are also found in an LC(20) homolog within the genome of Monosiga brevicollis, the closest living relative of metazoans. LC(20) diphosphorylation has been detected in mammalian vascular smooth muscle tissues in response to specific contractile stimuli and in pathophysiological situations associated with hypercontractility. LC(20) diphosphorylation has also been observed frequently in cultured cells where it activates force generation. Kinases such as ILK, ROCK, and ZIPK, therefore, are potential therapeutic targets in the treatment of, for example, cerebral vasospasm following subarachnoid hemorrhage and atherosclerosis.
Collapse
Affiliation(s)
- Michael P Walsh
- Smooth Muscle Research Group and Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta T2N 4N1, Canada.
| |
Collapse
|
39
|
Nepiyushchikh ZV, Chakraborty S, Wang W, Davis MJ, Zawieja DC, Muthuchamy M. Differential effects of myosin light chain kinase inhibition on contractility, force development and myosin light chain 20 phosphorylation of rat cervical and thoracic duct lymphatics. J Physiol 2011; 589:5415-29. [PMID: 21930597 DOI: 10.1113/jphysiol.2011.218446] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The intrinsic contractile activity of lymphatics varies in different regions of the body. We have previously shown that cervical lymphatics possess an inherently higher frequency but lower tone at a given pressure when compared to thoracic duct lymphatics. However, the molecular mechanisms modulating the contractile characteristics of these lymphatics are not well understood. Since myosin light chain 20 (MLC(20)) phosphorylation appears to underlie the tonic component of lymphatic contraction, we hypothesized that the thoracic duct would be more sensitive to the modulation of MLC(20) phosphorylation when compared to cervical lymphatics. To test our hypothesis, the contractile activities and MLC(20) phosphorylation of thoracic duct and cervical lymphatics were determined in the absence or presence of the specific myosin light chain kinase (MLCK) inhibitor ML-7 under both isobaric and isometric conditions. Addition of ML-7 at each concentration tested led to a decrease in tone in both vessel types. While ML-7 (10(-6) m) significantly reduced the phasic contraction frequency of cervical lymphatics, it completely stopped phasic contractions of thoracic duct at that concentration. Under isometric conditions the active peak and plateau components of tension were both significantly higher in thoracic duct compared to cervical lymphatics. ML-7 (10(-5) m) significantly decreased both the active peak and plateau tensions of thoracic duct, whereas only the active peak tension of cervical lymphatics was decreased. In thoracic duct MLC(20) di-phosphorylation, but not mono-phosphorylation, was significantly decreased with increasing transmural pressure, whereas in cervical vessels only at the higher pressures tested did MLC(20) di-phosphorylation decrease. ML-7 treatment of the thoracic duct caused a significant decrease in both the mono- and di-phosphorylated forms of MLC(20). However, in cervical vessels ML-7 treatment produced an increase in the mono-phosphorylated MLC(20) form while di-phosphorylated MLC(20) was significantly decreased. These data indicate that thoracic duct has an enhanced sensitivity to MLCK inhibition when compared to cervical lymphatics and while the status of the mono- and di-phosphorylation forms of MLC(20) affects both tonic and phasic components of lymphatic contractions, the pressure-dependent changes in tonic contractions are modulated by the status of the di-phosphorylation of MLC(20) in the lymphatics.
Collapse
Affiliation(s)
- Zhanna V Nepiyushchikh
- Department of Systems Biology and Translational Medicine, Cardiovascular Research Institute, Division of Lymphatic Biology, Texas A&M Health Science Center College of Medicine, 336 Reynolds Medical Building, College Station, TX 77843, USA
| | | | | | | | | | | |
Collapse
|
40
|
Borysova L, Shabir S, Walsh MP, Burdyga T. The importance of Rho-associated kinase-induced Ca2+ sensitization as a component of electromechanical and pharmacomechanical coupling in rat ureteric smooth muscle. Cell Calcium 2011; 50:393-405. [PMID: 21839512 DOI: 10.1016/j.ceca.2011.07.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Revised: 07/05/2011] [Accepted: 07/07/2011] [Indexed: 01/30/2023]
Abstract
Ureteric peristalsis, which occurs via alternating contraction and relaxation of ureteric smooth muscle, ensures the unidirectional flow of urine from the kidney to the bladder. Understanding of the molecular mechanisms underlying ureteric excitation-contraction coupling, however, is limited. To address these knowledge deficits, and in particular to test the hypothesis that Ca2+ sensitization via activation of the RhoA/Rho-associated kinase (ROK) pathway plays an important role in ureteric smooth muscle contraction, we carried out a thorough characterization of the electrical activity, Ca2+ signaling, MYPT1 (myosin targeting subunit of myosin light chain phosphatase, MLCP) and myosin regulatory light chain (LC20) phosphorylation, and force responses to membrane depolarization induced by KCl (electromechanical coupling) and carbachol (CCh) (pharmacomechanical coupling). The effects of ROK inhibition on these parameters were investigated. We conclude that the tonic, but not the phasic component of KCl- or CCh-induced ureteric smooth muscle contraction is highly dependent on ROK-catalyzed phosphorylation of MYPT1 at T855, leading to inhibition of MLCP and increased LC20 phosphorylation.
Collapse
Affiliation(s)
- Lyudmyla Borysova
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | | | | | | |
Collapse
|
41
|
Lim KM, Shin YS, Kang S, Noh JY, Kim K, Chung SM, Yun YP, Chung JH. Potentiation of vasoconstriction and pressor response by low concentration of monomethylarsonous acid (MMA(III)). Toxicol Lett 2011; 205:250-6. [PMID: 21708234 DOI: 10.1016/j.toxlet.2011.06.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Revised: 06/07/2011] [Accepted: 06/08/2011] [Indexed: 10/18/2022]
Abstract
A close link between arsenic exposure and hypertension has been well-established through many epidemiological reports, yet the mechanism underlying it remains unclear. Here we report that nanomolar concentrations of monomethylarsonous acid (MMA(III)), a toxic trivalent methylated arsenic metabolite, can potentiate agonist-induced vasoconstriction and pressor responses. In freshly isolated rat aortic ring, exposure to nanomolar MMA(III) (100-500 nM) potentiated phenylephrine (PE)-induced vasoconstriction while at higher concentrations (≥2.5 μM), suppression of vasoconstriction and apoptosis of vascular smooth muscle were observed. Potentiation of agonist-induced vasoconstriction was also observed with other contractile agonists and it was retained in endothelium-denuded aortic rings, suggesting that these events are agonist-independent and smooth muscle cell dependent. Interestingly, exposure to MMA(III) resulted in increased myosin light chain phosphorylation while PE-induced Ca2+ influx was not affected, reflecting that Ca2+ sensitization is involved. In line with this, MMA(III) enhanced agonist-induced activation of small GTPase RhoA, a key contributor to Ca2+ sensitization. Of note, treatment of MMA(III) to rats induced significantly higher pressor responses in vivo, demonstrating that this event can occur in vivo indeed. We believe that RhoA-mediated Ca2+ sensitization and the resultant potentiation of vasoconstriction by MMA(III) may shed light on arsenic-associated hypertension.
Collapse
Affiliation(s)
- Kyung-Min Lim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 151-742, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Role of myosin light chain kinase and myosin light chain phosphatase in the resistance arterial myogenic response to intravascular pressure. Arch Biochem Biophys 2011; 510:160-73. [DOI: 10.1016/j.abb.2011.02.024] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Revised: 02/24/2011] [Accepted: 02/28/2011] [Indexed: 12/19/2022]
|
43
|
Grassie ME, Moffat LD, Walsh MP, MacDonald JA. The myosin phosphatase targeting protein (MYPT) family: a regulated mechanism for achieving substrate specificity of the catalytic subunit of protein phosphatase type 1δ. Arch Biochem Biophys 2011; 510:147-59. [PMID: 21291858 DOI: 10.1016/j.abb.2011.01.018] [Citation(s) in RCA: 181] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Revised: 01/22/2011] [Accepted: 01/26/2011] [Indexed: 12/23/2022]
Abstract
The mammalian MYPT family consists of the products of five genes, denoted MYPT1, MYPT2, MBS85, MYPT3 and TIMAP, which function as targeting and regulatory subunits to confer substrate specificity and subcellular localization on the catalytic subunit of type 1δ protein serine/threonine phosphatase (PP1cδ). Family members share several conserved domains, including an RVxF motif for PP1c binding and several ankyrin repeats that mediate protein-protein interactions. MYPT1, MYPT2 and MBS85 contain C-terminal leucine zipper domains involved in dimerization and protein-protein interaction, whereas MYPT3 and TIMAP are targeted to membranes via a C-terminal prenylation site. All family members are regulated by phosphorylation at multiple sites by various protein kinases; for example, Rho-associated kinase phosphorylates MYPT1, MYPT2 and MBS85, resulting in inhibition of phosphatase activity and Ca(2+) sensitization of smooth muscle contraction. A great deal is known about MYPT1, the myosin targeting subunit of myosin light chain phosphatase, in terms of its role in the regulation of smooth muscle contraction and, to a lesser extent, non-muscle motile processes. MYPT2 appears to be the key myosin targeting subunit of myosin light chain phosphatase in cardiac and skeletal muscles. MBS85 most closely resembles MYPT2, but little is known about its physiological function. Little is also known about the physiological role of MYPT3, although it is likely to target myosin light chain phosphatase to membranes and thereby achieve specificity for substrates involved in regulation of the actin cytoskeleton. MYPT3 is regulated by phosphorylation by cAMP-dependent protein kinase. TIMAP appears to target PP1cδ to the plasma membrane of endothelial cells where it serves to dephosphorylate proteins involved in regulation of the actin cytoskeleton and thereby control endothelial barrier function. With such a wide range of regulatory targets, MYPT family members have been implicated in diverse pathological events, including hypertension, Parkinson's disease and cancer.
Collapse
Affiliation(s)
- Michael E Grassie
- Smooth Muscle Research Group, Department of Biochemistry and Molecular Biology, University of Calgary, AB, Canada
| | | | | | | |
Collapse
|