1
|
Lubomirov LT, Weber G, Schroeter M, Metzler D, Bust M, Korotkova T, Hescheler J, Todorov VT, Pfitzer G, Grisk O. Alanine mutation of the targeting subunit of the myosin phosphatase, MYPT1 at threonine 696 reduces cGMP responsiveness of mouse femoral arteries. Eur J Pharmacol 2025; 986:177133. [PMID: 39551336 DOI: 10.1016/j.ejphar.2024.177133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 11/07/2024] [Accepted: 11/13/2024] [Indexed: 11/19/2024]
Abstract
The femoral artery (FA) is the largest vessel in the hindlimb circulation and its proper tone regulation ensures adequate blood supply to muscle tissue. We investigated whether an alanine mutation of the targeting subunit of myosin-light-chain-phosphatase (MLCP), MYPT1, at threonine 696 (MYPT1-T696A/+), decisive for enzyme acivity, affects the responsiveness of young and old FAs (y-FAs and o-FAs) to activation of nitric-oxide/soluble-guanylate-cyclase/protein-kinase-G cascade (NO/sGC/PKG). Contractile responses of the vessels were measured by wire myography. Phosphorylation of the regulatory myosin-light-chain at serine 19 (MLC20-S19), the myosin-light-chain-phosphatase targeting subunit, MYPT1-T696, the PKG-sensitive site of MYPT1 at S695 (MYPT1-S695) and S668 (MYPT1-S668), and the regulatory phosphorylation of eNOS at S1177 (eNOS-S1177) were determined in arterial homogenates by Western blot. In FAs of all ages, the MYPT1-T696A-mutation did not alter vessel diameter and the contractile reactivity to the thromboxaneA2-analogue, U46619 and the RhoA kinase inhibitor, Y27632. In contrast, the mutation T696 into alanine attenuated the relaxing effect of exogenous NO (DEA-NONOate) in y-FAs. The effect of a direct sGC activation by cinaciguat was also attenuated in both age groups of MYPT1-T696A/+, but strongly in o-FA. The MYPT1-T696A-mutation also attenuated acetylcholine-induced relaxation, but only in o-FAs. Similary, the alanine mutation attenuated the acetylcholine effect on MLC20-S19- and MYPT1-T696 only in WT o-FAs. Interestingly, neither eNOS-S1177 nor the phosphorylation of the PKG phosphospecific sites, MYPT1-S695 and MYPT1-S668 were altered by MYPT1-T696A-mutation or aging. These findings suggest that the alanine mutation of MYPT1-T696 reduces the ability of the NO/cGMP/PKG-system to relax FAs in aging.
Collapse
Affiliation(s)
- Lubomir T Lubomirov
- Institute of Physiology, Brandenburg Medical School Theodor Fontane, Germany; Institute of Vegetative Physiology, Center of Physiology, University of Cologne, Germany; Vascular Biology Research Group (RenEVA), Research Institute, Medical University-Varna, Varna, Bulgaria; Institute of Physiology and Pathophysiology, Faculty of Health - School of Medicine, Biomedical Center for Education and Research (ZBAF), Witten/Herdecke University, Witten, Germany; Research Cluster, Molecular Mechanisms of Cardiovascular Diseases, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany.
| | - Greta Weber
- Institute of Physiology, Brandenburg Medical School Theodor Fontane, Germany
| | - Mechthild Schroeter
- Institute of Vegetative Physiology, Center of Physiology, University of Cologne, Germany
| | - Doris Metzler
- Institute of Vegetative Physiology, Center of Physiology, University of Cologne, Germany
| | - Maria Bust
- Institute of Vegetative Physiology, Center of Physiology, University of Cologne, Germany
| | - Tatiana Korotkova
- Institute of Vegetative Physiology, Center of Physiology, University of Cologne, Germany
| | - Jürgen Hescheler
- Institute of Neurophysiology, Center of Physiology, University of Cologne, Germany
| | - Vladimir T Todorov
- Institute of Physiology and Pathophysiology, Faculty of Health - School of Medicine, Biomedical Center for Education and Research (ZBAF), Witten/Herdecke University, Witten, Germany; Experimental Nephrology and Division of Nephrology, Department of Internal Medicine III, University Hospital and Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01307, Dresden, Germany
| | - Gabriele Pfitzer
- Institute of Vegetative Physiology, Center of Physiology, University of Cologne, Germany
| | - Olaf Grisk
- Institute of Physiology, Brandenburg Medical School Theodor Fontane, Germany; Research Cluster, Molecular Mechanisms of Cardiovascular Diseases, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
| |
Collapse
|
2
|
Han YS, Bandi R, Fogarty MJ, Sieck GC, Brozovich FV. Aging related decreases in NM myosin expression and contractility in a resistance vessel. Front Physiol 2024; 15:1411420. [PMID: 38808359 PMCID: PMC11130448 DOI: 10.3389/fphys.2024.1411420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 05/01/2024] [Indexed: 05/30/2024] Open
Abstract
Introduction: Vasodilatation in response to NO is a fundamental response of the vasculature, and during aging, the vasculature is characterized by an increase in stiffness and decrease in sensitivity to NO mediated vasodilatation. Vascular tone is regulated by the activation of smooth muscle and nonmuscle (NM) myosin, which are regulated by the activities of myosin light chain kinase (MLCK) and MLC phosphatase. MLC phosphatase is a trimeric enzyme with a catalytic subunit, myosin targeting subunit (MYPT1) and 20 kDa subunit of unknown function. Alternative mRNA splicing produces LZ+/LZ- MYPT1 isoforms and the relative expression of LZ+/LZ- MYPT1 determines the sensitivity to NO mediated vasodilatation. This study tested the hypothesis that aging is associated with changes in LZ+ MYPT1 and NM myosin expression, which alter vascular reactivity. Methods: We determined MYPT1 and NM myosin expression, force and the sensitivity of both endothelial dependent and endothelial independent relaxation in tertiary mesenteric arteries of young (6mo) and elderly (24mo) Fischer344 rats. Results: The data demonstrate that aging is associated with a decrease in both the expression of NM myosin and force, but LZ+ MYPT expression and the sensitivity to both endothelial dependent and independent vasodilatation did not change. Further, smooth muscle cell hypertrophy increases the thickness of the medial layer of smooth muscle with aging. Discussion: The reduction of NM myosin may represent an aging associated compensatory mechanism to normalize the stiffness of resistance vessels in response to the increase in media thickness observed during aging.
Collapse
Affiliation(s)
- Young Soo Han
- Departments of Physiology and Biomedical Engineering and Cardiovascular Diseases, Mayo Clinic, Rochester, MN, United States
| | - Rishiraj Bandi
- Departments of Physiology and Biomedical Engineering and Cardiovascular Diseases, Mayo Clinic, Rochester, MN, United States
| | - Matthew J Fogarty
- Departments of Physiology and Biomedical Engineering and Cardiovascular Diseases, Mayo Clinic, Rochester, MN, United States
| | - Gary C Sieck
- Departments of Physiology and Biomedical Engineering and Cardiovascular Diseases, Mayo Clinic, Rochester, MN, United States
| | - Frank V Brozovich
- Departments of Physiology and Biomedical Engineering and Cardiovascular Diseases, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
3
|
Lubomirov LT, Schroeter MM, Hasse V, Frohn M, Metzler D, Bust M, Pryymachuk G, Hescheler J, Grisk O, Chalovich JM, Smyth NR, Pfitzer G, Papadopoulos S. Dual thick and thin filament linked regulation of stretch- and L-NAME-induced tone in young and senescent murine basilar artery. Front Physiol 2023; 14:1099278. [PMID: 37057180 PMCID: PMC10088910 DOI: 10.3389/fphys.2023.1099278] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 03/09/2023] [Indexed: 03/30/2023] Open
Abstract
Stretch-induced vascular tone is an important element of autoregulatory adaptation of cerebral vasculature to maintain cerebral flow constant despite changes in perfusion pressure. Little is known as to the regulation of tone in senescent basilar arteries. We tested the hypothesis, that thin filament mechanisms in addition to smooth muscle myosin-II regulatory-light-chain-(MLC20)-phosphorylation and non-muscle-myosin-II, contribute to regulation of stretch-induced tone. In young BAs (y-BAs) mechanical stretch does not lead to spontaneous tone generation. Stretch-induced tone in y-BAs appeared only after inhibition of NO-release by L-NAME and was fully prevented by treatment with 3 μmol/L RhoA-kinase (ROK) inhibitor Y27632. L-NAME-induced tone was reduced in y-BAs from heterozygous mice carrying a point mutation of the targeting-subunit of the myosin phosphatase, MYPT1 at threonine696 (MYPT1-T696A/+). In y-BAs, MYPT1-T696A-mutation also blunted the ability of L-NAME to increase MLC20-phosphorylation. In contrast, senescent BAs (s-BAs; >24 months) developed stable spontaneous stretch-induced tone and pharmacological inhibition of NO-release by L-NAME led to an additive effect. In s-BAs the MYPT1-T696A mutation also blunted MLC20-phosphorylation, but did not prevent development of stretch-induced tone. In s-BAs from both lines, Y27632 completely abolished stretch- and L-NAME-induced tone. In s-BAs phosphorylation of non-muscle-myosin-S1943 and PAK1-T423, shown to be down-stream effectors of ROK was also reduced by Y27632 treatment. Stretch- and L-NAME tone were inhibited by inhibition of non-muscle myosin (NM-myosin) by blebbistatin. We also tested whether the substrate of PAK1 the thin-filament associated protein, caldesmon is involved in the regulation of stretch-induced tone in advanced age. BAs obtained from heterozygotes Cald1+/− mice generated stretch-induced tone already at an age of 20–21 months old BAs (o-BA). The magnitude of stretch-induced tone in Cald1+/− o-BAs was similar to that in s-BA. In addition, truncation of caldesmon myosin binding Exon2 (CaD-▵Ex2−/−) did not accelerate stretch-induced tone. Our study indicates that in senescent cerebral vessels, mechanisms distinct from MLC20 phosphorylation contribute to regulation of tone in the absence of a contractile agonist. While in y-and o-BA the canonical pathways, i.e., inhibition of MLCP by ROK and increase in pMLC20, predominate, tone regulation in senescence involves ROK regulated mechanisms, involving non-muscle-myosin and thin filament linked mechanisms involving caldesmon.
Collapse
Affiliation(s)
- Lubomir T. Lubomirov
- Center of Physiology, Institute of Vegetative Physiology, University of Cologne, Cologne, Germany
- Institute of Physiology, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
- Research Cluster, Molecular Mechanisms of Cardiovascular Diseases, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
- *Correspondence: Lubomir T. Lubomirov,
| | - Mechthild M. Schroeter
- Center of Physiology, Institute of Vegetative Physiology, University of Cologne, Cologne, Germany
- Center of Physiology, Institute of Neurophysiology, University of Cologne, Cologne, Germany
| | - Veronika Hasse
- Center of Physiology, Institute of Vegetative Physiology, University of Cologne, Cologne, Germany
| | - Marina Frohn
- Center of Physiology, Institute of Vegetative Physiology, University of Cologne, Cologne, Germany
| | - Doris Metzler
- Center of Physiology, Institute of Vegetative Physiology, University of Cologne, Cologne, Germany
| | - Maria Bust
- Center of Physiology, Institute of Vegetative Physiology, University of Cologne, Cologne, Germany
| | - Galyna Pryymachuk
- Institute of Anatomy, University of Cologne, Cologne, Germany
- Institute of Anatomy, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
| | - Jürgen Hescheler
- Center of Physiology, Institute of Neurophysiology, University of Cologne, Cologne, Germany
| | - Olaf Grisk
- Institute of Physiology, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
- Research Cluster, Molecular Mechanisms of Cardiovascular Diseases, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
| | - Joseph M. Chalovich
- Department of Biochemistry and Molecular Biology, Brody School of Medicine at East Carolina University, Greenville, NC, United States
| | - Neil R. Smyth
- Biological Sciences, Southampton General Hospital, University of Southampton, Southampton, United Kingdom
| | - Gabriele Pfitzer
- Center of Physiology, Institute of Vegetative Physiology, University of Cologne, Cologne, Germany
| | - Symeon Papadopoulos
- Center of Physiology, Institute of Neurophysiology, University of Cologne, Cologne, Germany
| |
Collapse
|
4
|
González-Herrera F, Clayton NS, Guzmán-Rivera D, Carrillo I, Castillo C, Catalán M, Anfossi R, Quintero-Pertuz H, Quilaqueo ME, Olea-Azar C, Rivera-Meza M, Kemmerling U, Ridley AJ, Vivar R, Maya JD. Statins change the cytokine profile in Trypanosoma cruzi-infected U937 macrophages and murine cardiac tissue through Rho-associated kinases inhibition. Front Immunol 2023; 13:1035589. [PMID: 36713380 PMCID: PMC9874148 DOI: 10.3389/fimmu.2022.1035589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 12/16/2022] [Indexed: 01/12/2023] Open
Abstract
Introduction Chronic Chagasic cardiomyopathy (CCC), caused by the protozoan Trypanosoma cruzi, is the most severe manifestation of Chagas disease.CCC is characterized by cardiac inflammation and fibrosis caused by a persistent inflammatory response. Following infection, macrophages secrete inflammatory mediators such as IL-1β, IL-6, and TNF-α to control parasitemia. Although this response contains parasite infection, it causes damage to the heart tissue. Thus, the use of immunomodulators is a rational alternative to CCC. Rho-associated kinase (ROCK) 1 and 2 are RhoA-activated serine/threonine kinases that regulate the actomyosin cytoskeleton. Both ROCKs have been implicated in the polarization of macrophages towards an M1 (pro-inflammatory) phenotype. Statins are FDA-approved lipid-lowering drugs that reduce RhoA signaling by inhibiting geranylgeranyl pyrophosphate (GGPP) synthesis. This work aims to identify the effect of statins on U937 macrophage polarization and cardiac tissue inflammation and its relationship with ROCK activity during T. cruzi infection. Methods PMA-induced, wild-type, GFP-, CA-ROCK1- and CA-ROCK2-expressing U937 macrophages were incubated with atorvastatin, or the inhibitors Y-27632, JSH-23, TAK-242, or C3 exoenzyme incubated with or without T. cruzi trypomastigotes for 30 min to evaluate the activity of ROCK and the M1 and M2 cytokine expression and secretion profiling. Also, ROCK activity was determined in T. cruzi-infected, BALB/c mice hearts. Results In this study, we demonstrate for the first time in macrophages that incubation with T. cruzi leads to ROCK activation via the TLR4 pathway, which triggers NF-κB activation. Inhibition of ROCKs by Y-27632 prevents NF-κB activation and the expression and secretion of M1 markers, as does treatment with atorvastatin. Furthermore, we show that the effect of atorvastatin on the NF-kB pathway and cytokine secretion is mediated by ROCK. Finally, statin treatment decreased ROCK activation and expression, and the pro-inflammatory cytokine production, promoting anti-inflammatory cytokine expression in chronic chagasic mice hearts. Conclusion These results suggest that the statin modulation of the inflammatory response due to ROCK inhibition is a potential pharmacological strategy to prevent cardiac inflammation in CCC.
Collapse
Affiliation(s)
- Fabiola González-Herrera
- Molecular and Clinical Pharmacology Program, Instituto de Ciencias Biomédicas, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Natasha S. Clayton
- School of Cellular and Molecular Medicine, Faculty of Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Daniela Guzmán-Rivera
- Escuela de Farmacia, Facultad de Medicina, Universidad Andrés Bello, Santiago, Chile
| | - Ileana Carrillo
- Molecular and Clinical Pharmacology Program, Instituto de Ciencias Biomédicas, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Christian Castillo
- Núcleo de Investigación Aplicada en Ciencias Veterinarias y Agronómicas, Facultad de Medicina Veterinaria y Agronomía, Universidad de Las Américas, Santiago, Chile
| | - Mabel Catalán
- Molecular and Clinical Pharmacology Program, Instituto de Ciencias Biomédicas, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Renatto Anfossi
- Molecular and Clinical Pharmacology Program, Instituto de Ciencias Biomédicas, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Helena Quintero-Pertuz
- Molecular and Clinical Pharmacology Program, Instituto de Ciencias Biomédicas, Faculty of Medicine, University of Chile, Santiago, Chile
| | - María Elena Quilaqueo
- Department of Chemical Pharmacology and Toxicology, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Claudio Olea-Azar
- Department of Inorganic and Analytical Chemistry, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Mario Rivera-Meza
- Department of Chemical Pharmacology and Toxicology, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Ulrike Kemmerling
- Integrative Biology Program, Instituto de Ciencias Biomédicas, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Anne J. Ridley
- School of Cellular and Molecular Medicine, Faculty of Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Raúl Vivar
- Molecular and Clinical Pharmacology Program, Instituto de Ciencias Biomédicas, Faculty of Medicine, University of Chile, Santiago, Chile,*Correspondence: Juan Diego Maya, ; Raúl Vivar,
| | - Juan Diego Maya
- Molecular and Clinical Pharmacology Program, Instituto de Ciencias Biomédicas, Faculty of Medicine, University of Chile, Santiago, Chile,*Correspondence: Juan Diego Maya, ; Raúl Vivar,
| |
Collapse
|
5
|
Kaneda T, Ifadotunnikmah F, Nugroho AE, Koshikawa S, Tadahiro S, Hirasawa Y, Morita H. Calofolic Acid-A from Calophyllum scriblitifolium Bark Has Vasorelaxant Activity via Indirect PKA Activation Caused by PI-3 Kinase Inhibition in Rat Vascular Smooth Muscle Cells. JOURNAL OF NATURAL PRODUCTS 2022; 85:2192-2198. [PMID: 35983865 DOI: 10.1021/acs.jnatprod.2c00502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Previously, we isolated 2R,3S,15R-calofolic acids (CAs) from Calophyllum scriblitifolium bark, which showed vasorelaxant activity on phenylephrine (PE)-precontracted rat aortic rings. Although the effect was suggested to be induced via an extracellular Ca2+-independent manner and mainly acts on vascular smooth muscle, the exact mechanism of action of CAs remained unclear. Thus, this study investigated the detailed mechanism of calofolic acid-A (CA-A) induced vasorelaxation in an aortic ring specimen using rat vascular smooth muscle cells (VSMCs). The levels of PE-induced phosphorylation on MLC Ser19 decreased in VSMCs pretreated with CA-A. CA-A also decreased the phosphorylation of MYPT1 Thr696 and MYPT1 Thr853. On the other hand, CA-A increased the PE-induced phosphorylation of MYPT1 Ser695 and MYPT1 Ser668, which are reported to be phosphorylated by a cAMP-dependent protein kinase (PKA). CA-A slightly increased PKA substrate phosphorylation in a concentration-dependent manner. Furthermore, CA-A enhanced isoproterenol (ISO)-induced cAMP accumulation and PKA substrate phosphorylation. Treatment with PI-3 kinase (PI3K) inhibitor, LY294002, enhanced ISO-induced cAMP accumulation and PKA substrate phosphorylation in the same manner as CA-A treatment. Furthermore, CA-A was found to directly inhibit PI3K enzyme activity in a dose-dependent manner. Taken together, the present study indicated that CA-A induces vasorelaxation through an indirectly activated PKA-MYPT1 pathway caused by inhibition of PI3K activity.
Collapse
Affiliation(s)
- Toshio Kaneda
- Faculty of Pharmaceutical Sciences, Hoshi University, Ebara 2-4-41, Shinagawa-ku, Tokyo 142-8501, Japan
| | - Farida Ifadotunnikmah
- Faculty of Pharmaceutical Sciences, Hoshi University, Ebara 2-4-41, Shinagawa-ku, Tokyo 142-8501, Japan
| | - Alfarius Eko Nugroho
- Faculty of Pharmaceutical Sciences, Hoshi University, Ebara 2-4-41, Shinagawa-ku, Tokyo 142-8501, Japan
| | - Sae Koshikawa
- Faculty of Pharmaceutical Sciences, Hoshi University, Ebara 2-4-41, Shinagawa-ku, Tokyo 142-8501, Japan
| | - Sasaki Tadahiro
- Faculty of Pharmaceutical Sciences, Hoshi University, Ebara 2-4-41, Shinagawa-ku, Tokyo 142-8501, Japan
| | - Yusuke Hirasawa
- Faculty of Pharmaceutical Sciences, Hoshi University, Ebara 2-4-41, Shinagawa-ku, Tokyo 142-8501, Japan
| | - Hiroshi Morita
- Faculty of Pharmaceutical Sciences, Hoshi University, Ebara 2-4-41, Shinagawa-ku, Tokyo 142-8501, Japan
| |
Collapse
|
6
|
Larson KF, Malik A, Brozovich FV. Aging and Heart Failure with Preserved Ejection Fraction. Compr Physiol 2022; 12:3813-3822. [PMID: 35950652 DOI: 10.1002/cphy.c210035] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Heart failure is a clinical syndrome characterized by the inability of the cardiovascular system to provide adequate cardiac output at normal filling pressures. This results in a clinical syndrome characterized by dyspnea, edema, and decreased exertional tolerance. Heart failure with preserved ejection fraction (HFpEF) is an increasingly common disease, and the incidence of HFpEF increases with age. There are a variety of factors which contribute to the development of HFpEF, including the presence of hypertension, diabetes, obesity, and other pro-inflammatory states. These comorbid conditions result in changes at the biochemical and cell signaling level which ultimately lead to a disease with a great deal of phenotypic heterogeneity. In general, the physiologic dysfunction of HFpEF is characterized by vascular stiffness, increased cardiac filling pressures, pulmonary hypertension, and impaired volume management. The normal and abnormal processes associated with aging serve as an accelerant in this process, resulting in the hypothesis that HFpEF represents a form of presbycardia. In this article, we aim to review the processes importance of aging in the development of HFpEF by examining the disease and its causes from the biochemical to physiologic level. © 2022 American Physiological Society. Compr Physiol 12: 1-10, 2022.
Collapse
Affiliation(s)
- Kathryn F Larson
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Awais Malik
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Frank V Brozovich
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Department of Physiology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
7
|
Saldanha PA, Bolanle IO, Palmer TM, Nikitenko LL, Rivero F. Complex Transcriptional Profiles of the PPP1R12A Gene in Cells of the Circulatory System as Revealed by In Silico Analysis and Reverse Transcription PCR. Cells 2022; 11:cells11152315. [PMID: 35954160 PMCID: PMC9367544 DOI: 10.3390/cells11152315] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 11/16/2022] Open
Abstract
The myosin light chain phosphatase target subunit 1 (MYPT1), encoded by the PPP1R12A gene, is a key component of the myosin light chain phosphatase (MLCP) protein complex. MYPT1 isoforms have been described as products of the cassette-type alternative splicing of exons E13, E14, E22, and E24. Through in silico analysis of the publicly available EST and mRNA databases, we established that PPP1R12A contains 32 exons (6 more than the 26 previously reported), of which 29 are used in 11 protein-coding transcripts. An in silico analysis of publicly available RNAseq data combined with validation by reverse transcription (RT)-PCR allowed us to determine the relative abundance of each transcript in three cell types of the circulatory system where MYPT1 plays important roles: human umbilical vein endothelial cells (HUVEC), human saphenous vein smooth muscle cells (HSVSMC), and platelets. All three cell types express up to 10 transcripts at variable frequencies. HUVECs and HSVSMCs predominantly express the full-length variant (58.3% and 64.3%, respectively) followed by the variant skipping E13 (33.7% and 23.1%, respectively), whereas in platelets the predominant variants are those skipping E14 (51.4%) and E13 (19.9%), followed by the full-length variant (14.4%). Variants including E24 account for 5.4% of transcripts in platelets but are rare (<1%) in HUVECs and HSVSMCs. Complex transcriptional profiles were also found across organs using in silico analysis of RNAseq data from the GTEx project. Our findings provide a platform for future studies investigating the specific (patho)physiological roles of understudied MYPT1 isoforms.
Collapse
|
8
|
Cui Y, Yang L, Liu X, Che C, Cheng J, Li P, Wen J, Yang Y. The decrease of MYPT1 is critical for impairment of NO-mediated vosodilation in mesenteric artery of the older spontaneously hypertensive rats. J Gerontol A Biol Sci Med Sci 2021; 77:424-432. [PMID: 34614147 DOI: 10.1093/gerona/glab290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Indexed: 11/13/2022] Open
Abstract
Nitric oxide (NO) mediated vasodilatation is a fundamental response of vasculature, however, the regulation of NO signaling pathway on resistance vessels in the elderly hypertension is still unclear. The 16-weeks-spontaneously hypertensive rats (SHR), the 18-months-SHR (OldSHR), and the age matched Wistar-Kyoto rats were used to study the changes of mesenteric resistance artery dilatation caused by sodium nitroprusside (SNP). After pre-vasoconstriction by Norepinephrine (NE), the response of endothelium-denuded mesenteric artery ring to SNP was observed, and the changes in vascular response after pharmacological interventions of key nodes in the NO/sGC/cGMP/PKG1α signaling pathway were observed as well. RNA sequencing and functional enrichment analyses were used to provide information for conducting validation experiments. Vasodilation of NO in OldSHR was decreased, which significantly correlated with the reduction of PKG-mediated effect. Functional enrichment analysis of RNA sequencing showed that genes encoding important proteins such as sGC and MYPT1 (protein phosphatase 1 regulatory subunit 12A) were downregulated in OldSHR. Molecular biology validation results showed that mRNA expression of both α and β subunits of sGC were reduced, while mRNA and protein expression of PKG1α were reduced in OldSHR. More importantly, the expression of MYPT1 and p S668-MYPT1 was significantly reduced in OldSHR, even under the treatment of SNP. The experiment also revealed an enhanced cAMP system in vasodilatory in hypertension, while this function completely lost in the elderly hypertension. Therefore, a NO-mediated decrease in vascular smooth muscle relaxation was found in the elderly hypertension. The dysfunction in cGMP-PKG signaling, in particular, the decreased p S668-MYPT1 was mechanistically involved.
Collapse
Affiliation(s)
- Yiqin Cui
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University
| | - Liju Yang
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University.,Department of Cardiology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Xiaoqin Liu
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University
| | - Chang Che
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University
| | - Jun Cheng
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University
| | - Pengyu Li
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University
| | - Jing Wen
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University
| | - Yan Yang
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University.,Department of Cardiology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
9
|
Lyle MA, Alabdaljabar MS, Han YS, Brozovich FV. The vasculature in HFpEF vs HFrEF: differences in contractile protein expression produce distinct phenotypes. Heliyon 2020; 6:e03129. [PMID: 31909283 PMCID: PMC6940630 DOI: 10.1016/j.heliyon.2019.e03129] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 11/21/2019] [Accepted: 12/23/2019] [Indexed: 12/14/2022] Open
Abstract
Both heart failure with reduced (HFrEF) and preserved (HFpEF) ejection fraction are associated with abnormalities of the vasculature, including a resting vasoconstriction and a decrease in sensitivity to nitric oxide (NO) mediated vasodilation. Vascular tone is controlled by the expression and activation of both smooth muscle (SM) and nonmuscle (NM) myosin, and NO mediated vasodilation is regulated by the expression of the leucine zipper positive (LZ+) isoform of the myosin targeting subunit (MYPT1) of myosin light chain phosphatase (MLCP). This study was designed to determine the expression of these contractile proteins in humans with HFrEF and HFpEF vs normal controls. We isolated tertiary mesenteric vessels from remnant biospecimens of patients undergoing partial or total colectomy at Mayo Clinic Rochester from August 2017 to December 2018, and examined the expression of MYPT1 and the LZ + MYPT1 isoform with immunoblots, while 2D SDS-PAGE was used to resolve the phosphorylated and nonphosphorylated regulatory light chains of NM and SM myosin. Our data show that NM myosin expression, as a percentage of total myosin, was 12 ± 3% (controls, n = 6), 7 ± 5% (HFpEF, n = 4) and 37 ± 18% (HFrEF, n = 5, p < 0.05). Total MYPT1 expression was significantly reduced (p < 0.05) in both HFpEF (70 ± 11%) and HFrEF (48 ± 6%); and in HFrEF, LZ + MYPT1 was also depressed (62 ± 19%, <0.05). These results demonstrate that HFrEF and HFpEF are distinct vascular entities, and the changes in protein expression contribute to the vascular abnormalities associated with these diseases. Further in HFpEF, the decrease in MYPT1 would explain why pharmacologic therapies that are designed to activate the NO/cGMP/PKG signaling pathway do not produce a clinical benefit.
Collapse
Affiliation(s)
- Melissa A. Lyle
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, 55905, USA
| | | | - Young Soo Han
- Department of Physiology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Frank V. Brozovich
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Physiology, Mayo Clinic, Rochester, MN, 55905, USA
| |
Collapse
|
10
|
Samson SC, Elliott A, Mueller BD, Kim Y, Carney KR, Bergman JP, Blenis J, Mendoza MC. p90 ribosomal S6 kinase (RSK) phosphorylates myosin phosphatase and thereby controls edge dynamics during cell migration. J Biol Chem 2019; 294:10846-10862. [PMID: 31138649 PMCID: PMC6635457 DOI: 10.1074/jbc.ra119.007431] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 05/08/2019] [Indexed: 12/25/2022] Open
Abstract
Cell migration is essential to embryonic development, wound healing, and cancer cell dissemination. Cells move via leading-edge protrusion, substrate adhesion, and retraction of the cell's rear. The molecular mechanisms by which extracellular cues signal to the actomyosin cytoskeleton to control these motility mechanics are poorly understood. The growth factor-responsive and oncogenically activated protein extracellular signal-regulated kinase (ERK) promotes motility by signaling in actin polymerization-mediated edge protrusion. Using a combination of immunoblotting, co-immunoprecipitation, and myosin-binding experiments and cell migration assays, we show here that ERK also signals to the contractile machinery through its substrate, p90 ribosomal S6 kinase (RSK). We probed the signaling and migration dynamics of multiple mammalian cell lines and found that RSK phosphorylates myosin phosphatase–targeting subunit 1 (MYPT1) at Ser-507, which promotes an interaction of Rho kinase (ROCK) with MYPT1 and inhibits myosin targeting. We find that by inhibiting the myosin phosphatase, ERK and RSK promote myosin II–mediated tension for lamella expansion and optimal edge dynamics for cell migration. These findings suggest that ERK activity can coordinately amplify both protrusive and contractile forces for optimal cell motility.
Collapse
Affiliation(s)
- Shiela C Samson
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112 and
| | - Andrew Elliott
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112 and
| | - Brian D Mueller
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112 and
| | - Yung Kim
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112 and
| | - Keith R Carney
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112 and
| | - Jared P Bergman
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112 and
| | - John Blenis
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115
| | - Michelle C Mendoza
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112 and; Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115.
| |
Collapse
|
11
|
LaFlamme A, Young KE, Lang I, Weiser DC. Alternative splicing of (ppp1r12a/mypt1) in zebrafish produces a novel myosin phosphatase targeting subunit. Gene 2018; 675:15-26. [PMID: 29960069 PMCID: PMC6123272 DOI: 10.1016/j.gene.2018.06.092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 06/07/2018] [Accepted: 06/26/2018] [Indexed: 01/04/2023]
Abstract
Myosin phosphatase is an evolutionarily conserved regulator of actomyosin contractility, comprised of a regulatory subunit (Mypt1), and a catalytic subunit (PP1). Zebrafish has become an ideal model organism for the study of the genetic and cell physiological role of the myosin phosphatase in morphogenesis and embryonic development. We identified and characterized a novel splice variant of Mypt1 (ppp1r12a-tv202) from zebrafish, which is widely expressed during early embryonic development. Importantly, mutant alleles and antisense morpholinos that have been used to demonstrate the important role of Mypt1 in early development, not only disrupt the longer splice variants, but also tv202. The protein product of ppp1r12a-tv202 (Mypt1-202) contains the PP1-binding N-terminus, but lacks the regulatory C-terminus, which contains two highly conserved inhibitory phosphorylation sites. We observed that the protein product of tv202 assembled a constitutively active myosin phosphatase uninhibited by kinases such as Zipk. Thus, we propose that Mypt1-202 plays an important role in maintaining baseline Mlc2 dephosphorylation and actomyosin relaxation during early zebrafish development.
Collapse
Affiliation(s)
- Andrew LaFlamme
- Department of Biological Sciences, University of the Pacific, Stockton, CA 95211, USA
| | - Kyle E Young
- Department of Biological Sciences, University of the Pacific, Stockton, CA 95211, USA
| | - Irene Lang
- Department of Biological Sciences, University of the Pacific, Stockton, CA 95211, USA
| | - Douglas C Weiser
- Department of Biological Sciences, University of the Pacific, Stockton, CA 95211, USA.
| |
Collapse
|
12
|
Sheehe JL, Bonev AD, Schmoker AM, Ballif BA, Nelson MT, Moon TM, Dostmann WR. Oxidation of cysteine 117 stimulates constitutive activation of the type Iα cGMP-dependent protein kinase. J Biol Chem 2018; 293:16791-16802. [PMID: 30206122 PMCID: PMC6204908 DOI: 10.1074/jbc.ra118.004363] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 09/04/2018] [Indexed: 12/22/2022] Open
Abstract
The type I cGMP-dependent protein kinase (PKG I) is an essential regulator of vascular tone. It has been demonstrated that the type Iα isoform can be constitutively activated by oxidizing conditions. However, the amino acid residues implicated in this phenomenon are not fully elucidated. To investigate the molecular basis for this mechanism, we studied the effects of oxidation using recombinant WT, truncated, and mutant constructs of PKG I. Using an in vitro assay, we observed that oxidation with hydrogen peroxide (H2O2) resulted in constitutive, cGMP-independent activation of PKG Iα. PKG Iα C42S and a truncation construct that does not contain Cys-42 (Δ53) were both constitutively activated by H2O2 In contrast, oxidation of PKG Iα C117S maintained its cGMP-dependent activation characteristics, although oxidized PKG Iα C195S did not. To corroborate these results, we also tested the effects of our constructs on the PKG Iα-specific substrate, the large conductance potassium channel (KCa 1.1). Application of WT PKG Iα activated by either cGMP or H2O2 increased the open probabilities of the channel. Neither cGMP nor H2O2 activation of PKG Iα C42S significantly increased channel open probabilities. Moreover, cGMP-stimulated PKG Iα C117S increased KCa 1.1 activity, but this effect was not observed under oxidizing conditions. Finally, we observed that PKG Iα C42S caused channel flickers, indicating dramatically altered KCa 1.1 channel characteristics compared with channels exposed to WT PKG Iα. Cumulatively, these results indicate that constitutive activation of PKG Iα proceeds through oxidation of Cys-117 and further suggest that the formation of a sulfur acid is necessary for this phenotype.
Collapse
Affiliation(s)
- Jessica L Sheehe
- From the Department of Pharmacology, Larner College of Medicine, and
| | - Adrian D Bonev
- From the Department of Pharmacology, Larner College of Medicine, and
| | - Anna M Schmoker
- the Department of Biology, University of Vermont, Burlington, Vermont 05405 and
| | - Bryan A Ballif
- the Department of Biology, University of Vermont, Burlington, Vermont 05405 and
| | - Mark T Nelson
- From the Department of Pharmacology, Larner College of Medicine, and
| | - Thomas M Moon
- the Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona 85721
| | | |
Collapse
|
13
|
Lyle MA, Brozovich FV. HFpEF, a Disease of the Vasculature: A Closer Look at the Other Half. Mayo Clin Proc 2018; 93:1305-1314. [PMID: 30064827 DOI: 10.1016/j.mayocp.2018.05.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 04/12/2018] [Accepted: 05/04/2018] [Indexed: 12/31/2022]
Abstract
Patients with heart failure are commonly divided into those with reduced ejection fraction (EF<40%) and those with preserved ejection fraction (HFpEF; EF>50%). For heart failure with reduced EF, a number of therapies have been found to improve patient morbidity and mortality, and treatment is guideline based. However for patients with HFpEF, no treatment has been found to have clinical benefit. To objectively assess treatments for HFpEF, a comprehensive PubMed literature search was performed using the terms HFpEF, heart failure, smooth muscle, myosin, myosin phosphatase, and PKG (up to December 31, 2017), with an unbiased focus on pathophysiology, cell signaling, and therapy. This review provides evidence that could explain the lack of clinical benefit in treating patients with HFpEF with sildenafil and long-acting nitrates. Furthermore, the review highlights the vascular abnormalities present in patients with HFpEF, and these abnormalities of the vasculature could potentially contribute to the pathophysiology of HFpEF. Thus, focusing on HFpEF as a vascular disease could result in the development of novel and effective treatment paradigms.
Collapse
Affiliation(s)
- Melissa A Lyle
- Department of Cadiovascular Diseases, Mayo Clinic College of Medicine and Science, Rochester, MN
| | - Frank V Brozovich
- Department of Cadiovascular Diseases, Mayo Clinic College of Medicine and Science, Rochester, MN.
| |
Collapse
|
14
|
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: 1.7] [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
|
15
|
Ghantous CM, Azrak Z, Rahman FA, Itani HA, Zeidan A. Assessment of Basilar Artery Reactivity in Stroke and Subarachnoid Hemorrhage Using Wire Myograph. Methods Mol Biol 2018; 1462:625-43. [PMID: 27604742 DOI: 10.1007/978-1-4939-3816-2_34] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Blood flow regulation of normal cerebral arteries is a critical and important factor to supply the brain tissue with nutrients and oxygen. Stroke insult results in a disruption or reduction in cerebral arteries' blood flow with subsequent brain tissue damage. Hemorrhagic stroke is one type of stroke and accounts for about 13 % of all of stroke insults. In this type of stroke, the cerebral artery breaks open and causes bleeding in or surrounding the brain. Subsequently, this bleeding causes blood vessels to constrict in a process called vasospasm, in which the vessels narrow and impede the blood flow to brain tissue. Hemorrhagic stroke is the major cause of prolonged constriction of cerebral arteries. This leads to partial brain damage and sometimes death in patients with aneurysmal subarachnoid hemorrhage. Among the key delicate techniques to assess small blood vessel functionality is the wire myograph, which can be utilized in several cerebral injury models including stroke. The wire myograph is a device that provides information about the reactivity, stiffness, and elasticity of small blood vessels under isometric conditions. In this book chapter, we describe the techniques involved in wire myography assessment and the different measures and parameters recorded; we describe the utility of this technique in evaluating the effects of subarachnoid hemorrhage on basilar artery sensitivity to different agonists.
Collapse
Affiliation(s)
- Crystal M Ghantous
- Department of Anatomy, Cell biology and Physiology, American University of Beirut, DTS-255, 11-0236, Beirut, 1107-2020, Lebanon
| | - Zeina Azrak
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon
| | - Farah Abdel Rahman
- Department of Anatomy, Cell biology and Physiology, American University of Beirut, DTS-255, 11-0236, Beirut, 1107-2020, Lebanon
| | - Hana A Itani
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Asad Zeidan
- Department of Anatomy, Cell biology and Physiology, American University of Beirut, DTS-255, 11-0236, Beirut, 1107-2020, Lebanon.
| |
Collapse
|
16
|
Lyle MA, Davis JP, Brozovich FV. Regulation of Pulmonary Vascular Smooth Muscle Contractility in Pulmonary Arterial Hypertension: Implications for Therapy. Front Physiol 2017; 8:614. [PMID: 28878690 PMCID: PMC5572347 DOI: 10.3389/fphys.2017.00614] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 08/09/2017] [Indexed: 12/23/2022] Open
Abstract
There are two primary components that produce pulmonary arterial hypertension (PAH); aberrant structural changes (smooth muscle cell proliferation, smooth muscle cell hypertrophy, and the deposition of matrix proteins within the media of pulmonary arterial vessels), and excess vasoconstriction. However, in PAH, the target and aim of all current therapeutic agents is to reduce the contractility of the pulmonary vasculature; prostaglandins, phosphodiesterase inhibitors, guanylate cyclase stimulators, endothelin antagonists, NO inhalation and Rho kinase inhibitors all influence signaling pathways in the pulmonary vascular smooth muscle to decrease vasoconstriction, and hence, pulmonary vascular resistance (PVR). This review will therefore primarily focus on discussing the signaling pathways regulating contractility in pulmonary vascular smooth muscle, the mechanism for current treatments, as well as highlighting potential targets for the development of novel therapies.
Collapse
Affiliation(s)
- Melissa A Lyle
- Department of Cardiovascular Diseases, Mayo ClinicRochester, MN, United States
| | - Jonathan P Davis
- Department of Physiology and Cell Biology, Ohio State UniversityColumbus, OH, United States
| | - Frank V Brozovich
- Department of Cardiovascular Diseases, Mayo ClinicRochester, MN, United States
| |
Collapse
|
17
|
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: 4] [Impact Index Per Article: 0.5] [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
|
18
|
Yin Y, Li Y, Pan J, Tang R, Zhu J, Qin Z, Xu X, Wang J. Expression of MYPT1, CPI-17 and MLC20 in ileum of neonatal mouse NEC model and its significance. Exp Ther Med 2017; 14:2221-2227. [PMID: 28962146 PMCID: PMC5609160 DOI: 10.3892/etm.2017.4783] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 07/04/2017] [Indexed: 11/06/2022] Open
Abstract
The present study determined the changes in the expression levels of MYPT1, CPI-17 and MLC20 in the ileum of mice with neonatal induced necrotizing enterocolitis (NEC) to provide a basis for a pathogenesis model that includes smooth muscle changes during NEC. A group of 7-day-old BALB/c mice were fed with formula (40 µl/g, 5 times/day) and given hypoxia treatments (5% O2 and 95% N2 for 10 min, twice daily) for 4 days to induce NEC and establish a mouse model. A control group of 7-day-old BALB/c mice were left with their mother for the duration of the treatment. After establishing the model, the two groups of mice were sacrificed, and the terminal ileum tissue was collected and subjected to western blot analysis and immunohistochemistry. The results showed the expression levels of MYPT1 and pMYPT1 in the ileum of the mice in the NEC group were lower than those in the control group (P<0.01). The levels of CPI17 and pCPI17 were higher in the NEC group compared with those in the control group. The expression level of MLC20 in NEC group was lower than that in the control group (P<0.01), but the level of pMLC20 in the NEC group was higher (P<0.05). The results of immunohistochemistry showed that the staining intensities of MYPT1, CPI-17 and MLC20 in the NEC group were lighter than those in the control group, and the proportion of positive cells was also lower in the NEC group (P<0.01). Taken together our results suggest that establishment of NEC is accompanied by changes in the protein levels of MYPT1 and pCPI-17, which can regulate smooth muscle contraction in the ileum.
Collapse
Affiliation(s)
- Yinyu Yin
- Department of General Surgery, Xuzhou Children's Hospital, Xuzhou, Jiangsu 221006, P.R. China
| | - Yiping Li
- Department of General Surgery, Children's Hospital of Soochow University, Suzhou, Jiangsu 215025, P.R. China
| | - Jian Pan
- Department of General Surgery, Children's Hospital of Soochow University, Suzhou, Jiangsu 215025, P.R. China
| | - Ruze Tang
- Department of General Surgery, Children's Hospital of Soochow University, Suzhou, Jiangsu 215025, P.R. China
| | - Jie Zhu
- Department of General Surgery, Children's Hospital of Soochow University, Suzhou, Jiangsu 215025, P.R. China
| | - Zhenfang Qin
- Department of General Surgery, Xuzhou Children's Hospital, Xuzhou, Jiangsu 221006, P.R. China
| | - Xiaobing Xu
- Department of General Surgery, Xuzhou Children's Hospital, Xuzhou, Jiangsu 221006, P.R. China
| | - Jian Wang
- Department of General Surgery, Children's Hospital of Soochow University, Suzhou, Jiangsu 215025, P.R. China
| |
Collapse
|
19
|
Filter JJ, Williams BC, Eto M, Shalloway D, Goldberg ML. Unfair competition governs the interaction of pCPI-17 with myosin phosphatase (PP1-MYPT1). eLife 2017; 6. [PMID: 28387646 PMCID: PMC5441869 DOI: 10.7554/elife.24665] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Accepted: 03/31/2017] [Indexed: 11/30/2022] Open
Abstract
The small phosphoprotein pCPI-17 inhibits myosin light-chain phosphatase (MLCP). Current models postulate that during muscle relaxation, phosphatases other than MLCP dephosphorylate and inactivate pCPI-17 to restore MLCP activity. We show here that such hypotheses are insufficient to account for the observed rapidity of pCPI-17 inactivation in mammalian smooth muscles. Instead, MLCP itself is the critical enzyme for pCPI-17 dephosphorylation. We call the mutual sequestration mechanism through which pCPI-17 and MLCP interact inhibition by unfair competition: MLCP protects pCPI-17 from other phosphatases, while pCPI-17 blocks other substrates from MLCP’s active site. MLCP dephosphorylates pCPI-17 at a slow rate that is, nonetheless, both sufficient and necessary to explain the speed of pCPI-17 dephosphorylation and the consequent MLCP activation during muscle relaxation. DOI:http://dx.doi.org/10.7554/eLife.24665.001
Collapse
Affiliation(s)
- Joshua J Filter
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| | - Byron C Williams
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| | - Masumi Eto
- Department of Molecular Physiology and Biophysics, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, United States
| | - David Shalloway
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| | - Michael L Goldberg
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| |
Collapse
|
20
|
Chettimada S, Joshi SR, Dhagia V, Aiezza A, Lincoln TM, Gupte R, Miano JM, Gupte SA. Vascular smooth muscle cell contractile protein expression is increased through protein kinase G-dependent and -independent pathways by glucose-6-phosphate dehydrogenase inhibition and deficiency. Am J Physiol Heart Circ Physiol 2016; 311:H904-H912. [PMID: 27521420 PMCID: PMC5114469 DOI: 10.1152/ajpheart.00335.2016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 07/06/2016] [Indexed: 11/22/2022]
Abstract
Homeostatic control of vascular smooth muscle cell (VSMC) differentiation is critical for contractile activity and regulation of blood flow. Recently, we reported that precontracted blood vessels are relaxed and the phenotype of VSMC is regulated from a synthetic to contractile state by glucose-6-phosphate dehydrogenase (G6PD) inhibition. In the current study, we investigated whether the increase in the expression of VSMC contractile proteins by inhibition and knockdown of G6PD is mediated through a protein kinase G (PKG)-dependent pathway and whether it regulates blood pressure. We found that the expression of VSMC-restricted contractile proteins, myocardin (MYOCD), and miR-1 and miR-143 are increased by G6PD inhibition or knockdown. Importantly, RNA-sequence analysis of aortic tissue from G6PD-deficient mice revealed uniform increases in VSMC-restricted genes, particularly those regulated by the MYOCD-serum response factor (SRF) switch. Conversely, expression of Krüppel-like factor 4 (KLF4) is decreased by G6PD inhibition. Interestingly, the G6PD inhibition-induced expression of miR-1 and contractile proteins was blocked by Rp-β-phenyl-1,N2-etheno-8-bromo-guanosine-3',5'-cyclic monophosphorothioate, a PKG inhibitor. On the other hand, MYOCD and miR-143 levels are increased by G6PD inhibition through a PKG-independent manner. Furthermore, blood pressure was lower in the G6PD-deficient compared with wild-type mice. Therefore, our results suggest that the expression of VSMC contractile proteins induced by G6PD inhibition occurs via PKG1α-dependent and -independent pathways.
Collapse
MESH Headings
- Animals
- Aorta/drug effects
- Aorta/metabolism
- Blotting, Western
- Cattle
- Chromatography, Liquid
- Contractile Proteins/drug effects
- Contractile Proteins/genetics
- Contractile Proteins/metabolism
- Cyclic GMP-Dependent Protein Kinase Type I/antagonists & inhibitors
- Cyclic GMP-Dependent Protein Kinase Type I/metabolism
- Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors
- Cyclic GMP-Dependent Protein Kinases/metabolism
- Gene Knockdown Techniques
- Glucosephosphate Dehydrogenase/antagonists & inhibitors
- Glucosephosphate Dehydrogenase/genetics
- Immunoprecipitation
- Kruppel-Like Factor 4
- Kruppel-Like Transcription Factors/drug effects
- Kruppel-Like Transcription Factors/genetics
- Kruppel-Like Transcription Factors/metabolism
- Mice
- MicroRNAs/drug effects
- MicroRNAs/genetics
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Nuclear Proteins/drug effects
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Polymerase Chain Reaction
- Rats
- Serum Response Factor/drug effects
- Serum Response Factor/genetics
- Serum Response Factor/metabolism
- Tandem Mass Spectrometry
- Trans-Activators/drug effects
- Trans-Activators/genetics
- Trans-Activators/metabolism
Collapse
Affiliation(s)
- Sukrutha Chettimada
- Biochemistry & Molecular Biology, University of South Alabama, Mobile, Alabama; Pharmacology, New York Medical College, Valhalla, New York
| | - Sachindra Raj Joshi
- Biochemistry & Molecular Biology, University of South Alabama, Mobile, Alabama; Pharmacology, New York Medical College, Valhalla, New York
| | - Vidhi Dhagia
- Pharmacology, New York Medical College, Valhalla, New York
| | - Alessandro Aiezza
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York; and
| | | | - Rakhee Gupte
- Biochemistry & Molecular Biology, University of South Alabama, Mobile, Alabama; Pharmacology, New York Medical College, Valhalla, New York
| | - Joseph M Miano
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York; and
| | - Sachin A Gupte
- Biochemistry & Molecular Biology, University of South Alabama, Mobile, Alabama; Pharmacology, New York Medical College, Valhalla, New York
| |
Collapse
|
21
|
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: 337] [Impact Index Per Article: 37.4] [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
|
22
|
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: 1.7] [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
|
23
|
Dippold RP, Fisher SA. Myosin phosphatase isoforms as determinants of smooth muscle contractile function and calcium sensitivity of force production. Microcirculation 2015; 21:239-48. [PMID: 24112301 DOI: 10.1111/micc.12097] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 09/25/2013] [Indexed: 12/14/2022]
Abstract
The dephosphorylation of myosin by the MP causes smooth muscle relaxation. MP is also a key target of signals that regulate vascular tone and thus blood flow and pressure. Here, we review studies from the past two decades that support the hypothesis that the regulated expression of MP subunits is a critical determinant of smooth muscle responses to constrictor and dilator signals. In particular, the highly regulated splicing of the regulatory subunit Mypt1 Exon 24 is proposed to tune sensitivity to NO/cGMP-mediated relaxation. The regulated transcription of the MP inhibitory subunit CPI-17 is proposed to determine sensitivity to agonist-mediated constriction. The expression of these subunits is specific in the microcirculation and varies in developmental and disease contexts. To date, the relationship between MP subunit expression and vascular function in these different contexts is correlative; confirmation of the hypothesis will require the generation of genetically engineered mice to test the role of MP subunits and their isoforms in the specificity of vascular smooth muscle responses to constrictor and dilator signals.
Collapse
Affiliation(s)
- Rachael P Dippold
- Department of Medicine (Cardiology), University of Maryland Baltimore, Baltimore, Maryland, USA
| | | |
Collapse
|
24
|
Chen CP, Chen X, Qiao YN, Wang P, He WQ, Zhang CH, Zhao W, Gao YQ, Chen C, Tao T, Sun J, Wang Y, Gao N, Kamm KE, Stull JT, Zhu MS. In vivo roles for myosin phosphatase targeting subunit-1 phosphorylation sites T694 and T852 in bladder smooth muscle contraction. J Physiol 2014; 593:681-700. [PMID: 25433069 DOI: 10.1113/jphysiol.2014.283853] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 11/18/2014] [Indexed: 01/12/2023] Open
Abstract
KEY POINTS Force production and maintenance in smooth muscle is largely controlled by myosin regulatory light chain (RLC) phosphorylation, which relies on a balance between Ca(2+)/calmodulin-dependent myosin light chain kinase (MLCK) and myosin light chain phosphatase (MLCP) activities. MYPT1 is the regulatory subunit of MLCP that biochemically inhibits MLCP activity via T694 or T852 phosphorylation in vitro. Here we separately investigated the contribution of these two phosphorylation sites in bladder smooth muscles by establishing two single point mutation mouse lines, T694A and T852A, and found that phosphorylation of MYPT1 T694, but not T852, mediates force maintenance via inhibition of MLCP activity and enhancement of RLC phosphorylation in vivo. Our findings reveal the role of MYPT1 T694/T852 phosphorylation in vivo in regulation of smooth muscle contraction. ABSTRACT Force production and maintenance in smooth muscle is largely controlled by different signalling modules that fine tune myosin regulatory light chain (RLC) phosphorylation, which relies on a balance between Ca(2+)/calmodulin-dependent myosin light chain kinase (MLCK) and myosin light chain phosphatase (MLCP) activities. To investigate the regulation of MLCP activity in vivo, we analysed the role of two phosphorylation sites on MYPT1 (regulatory subunit of MLCP) that biochemically inhibit MLCP activity in vitro. MYPT1 is constitutively phosphorylated at T694 by unidentified kinases in vivo, whereas the T852 site is phosphorylated by RhoA-associated protein kinase (ROCK). We established two mouse lines with alanine substitution of T694 or T852. Isolated bladder smooth muscle from T852A mice displayed no significant changes in RLC phosphorylation or force responses, but force was inhibited with a ROCK inhibitor. In contrast, smooth muscles containing the T694A mutation showed a significant reduction of force along with reduced RLC phosphorylation. The contractile responses of T694A mutant smooth muscle were also independent of ROCK activation. Thus, phosphorylation of MYPT1 T694, but not T852, is a primary mechanism contributing to inhibition of MLCP activity and enhancement of RLC phosphorylation in vivo. The constitutive phosphorylation of MYPT1 T694 may provide a mechanism for regulating force maintenance of smooth muscle.
Collapse
Affiliation(s)
- Cai-Ping Chen
- Model Animal Research Center and MOE Key Laboratory of Animal Models of Disease, Nanjing University, Nanjing, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Mahavadi S, Nalli A, Al-Shboul O, Murthy KS. Inhibition of MLC20 phosphorylation downstream of Ca2+ and RhoA: A novel mechanism involving phosphorylation of myosin phosphatase interacting protein (M-RIP) by PKG and stimulation of MLC phosphatase activity. Cell Biochem Biophys 2014; 68:1-8. [PMID: 23723008 DOI: 10.1007/s12013-013-9677-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Previous studies have shown that cGMP-dependent protein kinase (PKG) act on several targets in the contractile pathway to reduce intracellular Ca(2+) and/or augment RhoA-regulated myosin light chain phosphatase (MLCP) activity and cause muscle relaxation. Recent studies have identified a novel protein M-RIP that associates with MYPT1, the regulatory subunit of MLCP. Herein, we examine whether PKG enhance MLCP activity downstream of Ca(2+) and RhoA via phosphorylation of M-RIP in gastric smooth muscle cells. Treatment of permeabilized muscle cells with 10 μM Ca(2+) caused an increase in MLC20 phosphorylation and muscle contraction, but had no effect on Rho kinase activity. Activators of PKG (GSNO or cGMP) decreased MLC20 phosphorylation and contraction in response to 10 μM Ca(2+), implying existence of inhibitory mechanism independent of Ca(2+) and RhoA. The effect of PKG on Ca(2+)-induced MLC20 phosphorylation was attenuated by M-RIP siRNA. Both GSNO and 8-pCPT-cGMP induced phosphorylation of M-RIP; phosphorylation was accompanied by an increase in the association of M-RIP with MYPT1 and MLCP activity. Taken together, these results provide evidence that PKG induces phosphorylation of M-RIP and enhances its association with MYPT1 to augment MLCP activity and MLC20 dephosphorylation and inhibits muscle contraction, downstream of Ca(2+)- or RhoA-dependent pathways.
Collapse
Affiliation(s)
- Sunila Mahavadi
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, 23298-0711, USA
| | | | | | | |
Collapse
|
26
|
Yuen SL, Ogut O, Brozovich FV. Differential phosphorylation of LZ+/LZ- MYPT1 isoforms regulates MLC phosphatase activity. Arch Biochem Biophys 2014; 562:37-42. [PMID: 25168281 DOI: 10.1016/j.abb.2014.08.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 08/12/2014] [Accepted: 08/17/2014] [Indexed: 02/05/2023]
Abstract
The vascular response to NO is due, in part, to a Ca(2+) independent activation of myosin light chain (MLC) phosphatase, a trimeric enzyme of 20kDa, 38kDa catalytic and 110-130kDa myosin targeting (MYPT1) subunits. Alternative mRNA splicing produces MYPT1 isoforms that differ by the presence or absence of a central insert (CI) and a leucine zipper (LZ), and the presence of a LZ+ MYPT1 isoform is important for protein kinase G (PKG) mediated activation of MLC phosphatase. This study was designed to determine the molecular basis for the differential sensitivity of the vasculature to NO. Our results demonstrate that the presence of the MYPT1 LZ domain is required for PKG to both phosphorylate MYPT1 at S668 and activate MLC phosphatase. Further for LZ+ MYPT1 isoforms, an S668A MYPT1 mutation prevents the PKG mediated, Ca(2+) independent activation of MLC phosphatase. These data demonstrate that differential PKG mediated S668 phosphorylation of LZ+/LZ- MYPT1 isoforms could be important for determining the diversity in the sensitivity of the vasculature to NO mediated vasodilatation. Thus, the relative expression of LZ+/LZ- MYPT1 isoforms, in part, defines the vascular response to NO and NO based vasodilators, and therefore, plays a role in the regulation of vascular tone in both health and disease.
Collapse
Affiliation(s)
- Samantha L Yuen
- Division of Cardiovascular Diseases, Mayo Medical School, Rochester, MN 55905, USA
| | - Ozgur Ogut
- Division of Cardiovascular Diseases, Mayo Medical School, Rochester, MN 55905, USA
| | - Frank V Brozovich
- Division of Cardiovascular Diseases, Mayo Medical School, Rochester, MN 55905, USA.
| |
Collapse
|
27
|
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: 5.0] [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
|
28
|
Turner SR, MacDonald JA. Novel Contributions of the Smoothelin-like 1 Protein in Vascular Smooth Muscle Contraction and its Potential Involvement in Myogenic Tone. Microcirculation 2014; 21:249-58. [DOI: 10.1111/micc.12108] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 11/04/2013] [Indexed: 12/19/2022]
Affiliation(s)
- Sara R. Turner
- The Smooth Muscle Research Group at the Libin Cardiovascular Institute of Alberta; Department of Biochemistry & Molecular Biology; University of Calgary; Calgary Alberta Canada
| | - Justin A. MacDonald
- The Smooth Muscle Research Group at the Libin Cardiovascular Institute of Alberta; Department of Biochemistry & Molecular Biology; University of Calgary; Calgary Alberta Canada
| |
Collapse
|
29
|
Regulation of vascular function on posttranscriptional level. THROMBOSIS 2013; 2013:948765. [PMID: 24288605 PMCID: PMC3833109 DOI: 10.1155/2013/948765] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 09/17/2013] [Indexed: 11/17/2022]
Abstract
Posttranscriptional control of gene expression is crucial for regulating plurality of proteins and functional plasticity of the proteome under (patho)physiologic conditions. Alternative splicing as well as micro (mi)RNA-mediated mechanisms play an important role for the regulation of protein expression on posttranscriptional level. Both alternative splicing and miRNAs were shown to influence cardiovascular functions, such as endothelial thrombogenicity and the vascular tone, by regulating the expression of several vascular proteins and their isoforms, such as Tissue Factor (TF) or the endothelial nitric oxide synthase (eNOS). This review will summarize and discuss the latest findings on the (patho)physiologic role of alternative splicing processes as well as of miRNAs on modulation of vascular functions, such as coagulation, thrombosis, and regulation of the vascular tone.
Collapse
|
30
|
The role of pulmonary vascular contractile protein expression in pulmonary arterial hypertension. J Mol Cell Cardiol 2013; 65:147-55. [PMID: 24161910 DOI: 10.1016/j.yjmcc.2013.10.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 09/30/2013] [Accepted: 10/15/2013] [Indexed: 12/13/2022]
Abstract
Pulmonary arterial hypertension (PAH) is associated with refractory vasoconstriction and impaired NO-mediated vasodilatation of the pulmonary vasculature. Vascular tone is regulated by light chain (LC) phosphorylation of both nonmuscle (NM) and smooth muscle (SM) myosins, which are determined by the activities of MLC kinase and MLC phosphatase. Further, NO mediated vasodilatation requires the expression of a leucine zipper positive (LZ+) isoform of the myosin targeting subunit (MYPT1) of MLC phosphatase. The objective of this study was to define contractile protein expression in the pulmonary arterial vasculature and vascular reactivity in PAH. In severe PAH, compared to controls, relative LZ+MYPT1 expression was decreased (100 ± 14% vs. 60 ± 6%, p<0.05, n=7-8), and NM myosin expression was increased (1 5 ± 4% vs. 53 ± 5% of total myosin, p<0.05, n=4-6). These changes in contractile protein expression should alter vascular reactivity; following activation with Ang II, force activation and relaxation were slowed, and sustained force was increased. Further, the sensitivity to ACh-mediated relaxation was reduced. These results demonstrate that changes in the pulmonary arterial SM contractile protein expression may participate in the molecular mechanism producing both the resting vasoconstriction and the decreased sensitivity to NO-mediated vasodilatation associated with PAH.
Collapse
|
31
|
Dordea AC, Sweeney M, Taggart J, Lartey J, Wessel H, Robson SC, Taggart MJ. Differential vasodilation of human placental and myometrial arteries related to myofilament Ca(2+)-desensitization and the expression of Hsp20 but not MYPT1. Mol Hum Reprod 2013; 19:727-36. [PMID: 23775458 DOI: 10.1093/molehr/gat045] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Endothelial-dependent regulation of vascular tone occurs in part via protein kinase G1α-mediated changes in smooth muscle myofilament sensitivity to Ca(2+). Tissue-specific differences in PKG-dependent relaxation have been attributed to altered expression of myofilament-associated proteins that are substrates for PKG binding. These include the alternative splicing of the myosin targeting subunit (MYPT1) of myosin light chain phosphatase to yield leucine zipper positive (LZ(+)) and negative (LZ(-)) isovariants, with the former being required for PKG-mediated relaxation, and/or altered expressions of telokin, vasodilator-stimulated phosphoprotein (VASP) or heat shock protein Hsp20. During human pregnancy the uterine and placental circulations remain distinct entities and, as such, their mechanisms of vascular tone regulation may differ. Indeed, the sensitivity of myometrial arteries to endothelial-dependent agonists has been suggested to be greater than that of placental arteries. We tested the hypothesis that this was related to tissue-specific changes in PKG-mediated myofilament Ca(2+)-desensitization and/or the expressions of PKG-interacting myofilament-associated proteins. Permeabilized human placental and myometrial arteries were constricted with maximal activating Ca(2+) (pCa 4.5), or sub-maximal Ca(2+) (pCa 6.7) and the thrombane mimetic U46619, and exposed to 8-Br-cGMP. In each case, relaxation was significantly greater in myometrial arteries (e.g. relaxation in pCa 4.5 to 8-Br-cGMP was 49 ± 9.7%, n = 7) than placental arteries (relaxation of 23 ± 6.6%, n = 6, P < 0.05). MYPT1 protein levels, or MYPT1 LZ(+)/LZ(-) mRNA ratios, were similar for both artery types. Of other proteins examined, only Hsp20 expression was significantly elevated in myometrial arteries than placental arteries. These results demonstrate that the reduced human placental artery relaxation to PKG stimulation lies partly at the level of myofilament (de)activation and may be related to a lower expression of Hsp20 than in myometrial arteries.
Collapse
Affiliation(s)
- A C Dordea
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | | | | | | | | | | | | |
Collapse
|
32
|
Bhetwal BP, An C, Baker SA, Lyon KL, Perrino BA. Impaired contractile responses and altered expression and phosphorylation of Ca(2+) sensitization proteins in gastric antrum smooth muscles from ob/ob mice. J Muscle Res Cell Motil 2013; 34:137-49. [PMID: 23576331 DOI: 10.1007/s10974-013-9341-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 03/27/2013] [Indexed: 12/21/2022]
Abstract
Diabetic gastroparesis is a common complication of diabetes, adversely affecting quality of life with symptoms of abdominal discomfort, nausea, and vomiting. The pathogenesis of this complex disorder is not well understood, involving abnormalities in the extrinsic and enteric nervous systems, interstitial cells of Cajal (ICCs), smooth muscles and immune cells. The ob/ob mouse model of obesity and diabetes develops delayed gastric emptying, providing an animal model for investigating how gastric smooth muscle dysfunction contributes to the pathophysiology of diabetic gastroparesis. Although ROCK2, MYPT1, and CPI-17 activities are reduced in intestinal motility disorders, their functioning has not been investigated in diabetic gastroparesis. We hypothesized that reduced expression and phosphorylation of the myosin light chain phosphatase (MLCP) inhibitory proteins MYPT1 and CPI-17 in ob/ob gastric antrum smooth muscles could contribute to the impaired antrum smooth muscle function of diabetic gastroparesis. Spontaneous and carbachol- and high K(+)-evoked contractions of gastric antrum smooth muscles from 7 to 12 week old male ob/ob mice were reduced compared to age- and strain-matched controls. There were no differences in spontaneous and agonist-evoked intracellular Ca(2+) transients and myosin light chain kinase expression. The F-actin:G-actin ratios were similar. Rho kinase 2 (ROCK2) expression was decreased at both ages. Basal and agonist-evoked MYPT1 and myosin light chain 20 phosphorylation, but not CPI-17 phosphorylation, was reduced compared to age-matched controls. These findings suggest that reduced MLCP inhibition due to decreased ROCK2 phosphorylation of MYPT1 in gastric antrum smooth muscles contributes to the antral dysmotility of diabetic gastroparesis.
Collapse
Affiliation(s)
- Bhupal P Bhetwal
- Department of Physiology & Cell Biology, Center of Biomedical Research Excellence, University of Nevada School of Medicine, CMM 203E-MS 0575, 1664 N Virginia St, Reno, NV 89557, USA
| | | | | | | | | |
Collapse
|
33
|
The p90 ribosomal S6 kinase (RSK) is a mediator of smooth muscle contractility. PLoS One 2013; 8:e58703. [PMID: 23516539 PMCID: PMC3596281 DOI: 10.1371/journal.pone.0058703] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 02/05/2013] [Indexed: 11/19/2022] Open
Abstract
In the canonical model of smooth muscle (SM) contraction, the contractile force is generated by phosphorylation of the myosin regulatory light chain (RLC20) by the myosin light chain kinase (MLCK). Moreover, phosphorylation of the myosin targeting subunit (MYPT1) of the RLC20 phosphatase (MLCP) by the RhoA-dependent ROCK kinase, inhibits the phosphatase activity and consequently inhibits dephosphorylation of RLC20 with concomitant increase in contractile force, at constant intracellular [Ca2+]. This pathway is referred to as Ca2+-sensitization. There is, however, emerging evidence suggesting that additional Ser/Thr kinases may contribute to the regulatory pathways in SM. Here, we report data implicating the p90 ribosomal S6 kinase (RSK) in SM contractility. During both Ca2+- and agonist (U46619) induced SM contraction, RSK inhibition by the highly selective compound BI-D1870 (which has no effect on MLCK or ROCK) resulted in significant suppression of contractile force. Furthermore, phosphorylation levels of RLC20 and MYPT1 were both significantly decreased. Experiments involving the irreversible MLCP inhibitor microcystin-LR, in the absence of Ca2+, revealed that the decrease in phosphorylation levels of RLC20 upon RSK inhibition are not due solely to the increase in the phosphatase activity, but reflect direct or indirect phosphorylation of RLC20 by RSK. Finally, we show that agonist (U46619) stimulation of SM leads to activation of extracellular signal-regulated kinases ERK1/2 and PDK1, consistent with a canonical activation cascade for RSK. Thus, we demonstrate a novel and important physiological function of the p90 ribosomal S6 kinase, which to date has been typically associated with the regulation of gene expression.
Collapse
|
34
|
Butler T, Paul J, Europe-Finner N, Smith R, Chan EC. Role of serine-threonine phosphoprotein phosphatases in smooth muscle contractility. Am J Physiol Cell Physiol 2013; 304:C485-504. [PMID: 23325405 DOI: 10.1152/ajpcell.00161.2012] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The degree of phosphorylation of myosin light chain 20 (MLC20) is a major determinant of force generation in smooth muscle. Myosin phosphatases (MPs) contain protein phosphatase (PP) 1 as catalytic subunits and are the major enzymes that dephosphorylate MLC20. MP regulatory targeting subunit 1 (MYPT1), the main regulatory subunit of MP in all smooth muscles, is a key convergence point of contractile and relaxatory pathways. Combinations of regulatory mechanisms, including isoform splicing, multiple phosphorylation sites, and scaffolding proteins, modulate MYPT1 activity with tissue and agonist specificities to affect contraction and relaxation. Other members of the PP1 family that do not target myosin, as well as PP2A and PP2B, dephosphorylate a range of proteins that affect smooth muscle contraction. This review discusses the role of phosphatases in smooth muscle contractility with a focus on MYPT1 in uterine smooth muscle. Myometrium shares characteristics of vascular and other visceral smooth muscles yet, during healthy pregnancy, undergoes hypertrophy, hyperplasia, quiescence, and labor as physiological processes. Myometrium presents an accessible model for the study of normal and pathological smooth muscle function, and a better understanding of myometrial physiology may allow the development of novel therapeutics for the many disorders of myometrial physiology from preterm labor to dysmenorrhea.
Collapse
Affiliation(s)
- Trent Butler
- Mothers and Babies Research Centre, Faculty of Health, University of Newcastle, Callaghan, NSW 2308, Australia
| | | | | | | | | |
Collapse
|
35
|
Fu K, Mende Y, Bhetwal BP, Baker S, Perrino BA, Wirth B, Fisher SA. Tra2β protein is required for tissue-specific splicing of a smooth muscle myosin phosphatase targeting subunit alternative exon. J Biol Chem 2012; 287:16575-85. [PMID: 22437831 DOI: 10.1074/jbc.m111.325761] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Alternative splicing of the smooth muscle myosin phosphatase targeting subunit (Mypt1) exon 23 (E23) is tissue-specific and developmentally regulated and, thus, an attractive model for the study of smooth muscle phenotypic specification. We have proposed that Tra2β functions as a tissue-specific activator of Mypt1 E23 splicing on the basis of concordant expression patterns and Tra2β activation of Mypt1 E23 mini-gene splicing in vitro. In this study we examined the relationship between Tra2β and Mypt1 E23 splicing in vivo in the mouse. Tra2β was 2- to 5-fold more abundant in phasic smooth muscle tissues, such as the portal vein, small intestine, and small mesenteric artery, in which Mypt1 E23 is predominately included as compared with the tonic smooth muscle tissues, such as the aorta and inferior vena cava, in which Mypt1 E23 is predominately skipped. Tra2β was up-regulated in the small intestine postnatally, concordant with a switch to Mypt1 E23 splicing. Targeting of Tra2β in smooth muscle cells using SM22α-Cre caused a substantial reduction in Mypt1 E23 inclusion specifically in the intestinal smooth muscle of heterozygotes, indicating sensitivity to Tra2β gene dosage. The switch to the Mypt1 E23 skipped isoform coding for the C-terminal leucine zipper motif caused increased sensitivity of the muscle to the relaxant effects of 8-Br-cyclic guanosine monophosphate (cGMP). We conclude that Tra2β is necessary for the tissue-specific splicing of Mypt1 E23 in the phasic intestinal smooth muscle. Tra2β, by regulating the splicing of Mypt1 E23, sets the sensitivity of smooth muscle to cGMP-mediated relaxation.
Collapse
Affiliation(s)
- Kang Fu
- Department of Medicine (Cardiology), Case Western Reserve University, Cleveland, Ohio 44106, USA
| | | | | | | | | | | | | |
Collapse
|