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
|
Wang Y, Jiang ZH, Zhou YW, Qiu TT, Wang H, Zhu MS, Chen X, Zhang XN. Gallbladder dysfunction caused by MYPT1 ablation triggers cholestasis-induced hepatic fibrosis in mice. Hepatol Commun 2024; 8:e0473. [PMID: 38934703 PMCID: PMC11213606 DOI: 10.1097/hc9.0000000000000473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 04/19/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND The incidence of gallbladder diseases is as high as 20%, but whether gallbladder diseases contribute to hepatic disorders remains unknown. METHODS Here, we established an animal model of gallbladder dysfunction and assessed the role of a diseased gallbladder in cholestasis-induced hepatic fibrosis (CIHF). RESULTS Mice with smooth muscle-specific deletion of Mypt1, the gene encoding the main regulatory subunit of myosin light chain phosphatase (myosin phosphatase target subunit 1 [MYPT1]), had apparent dysfunction of gallbladder motility. This dysfunction was evidenced by abnormal contractile responses, namely, inhibited cholecystokinin 8-mediated contraction and nitric oxide-resistant relaxation. As a consequence, the gallbladder displayed impaired bile filling and biliary tract dilation comparable to the alterations in CIHF. Interestingly, the mutant animals also displayed CIHF features, including necrotic loci by the age of 1 month and subsequently exhibited progressive fibrosis and hyperplastic/dilated bile ducts. This pathological progression was similar to the phenotypes of the animal model with bile duct ligation and patients with CIHF. The characteristic biomarker of CIHF, serum alkaline phosphatase activity, was also elevated in the mice. Moreover, we observed that the myosin phosphatase target subunit 1 protein level was able to be regulated by several reagents, including lipopolysaccharide, exemplifying the risk factors for gallbladder dysfunction and hence CIHF. CONCLUSIONS We propose that gallbladder dysfunction caused by myosin phosphatase target subunit 1 ablation is sufficient to induce CIHF in mice, resulting in impairment of the bile transport system.
Collapse
Affiliation(s)
- Ye Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing, China
| | - Zhi-Hui Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing, China
| | - Yu-Wei Zhou
- Jiangsu Key Laboratory of Molecular Medicine, Department of Otolaryngology Head and Neck Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Tian-Tian Qiu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing, China
| | - Han Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing, China
| | - Min-Sheng Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing, China
| | - Xin Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing, China
| | - Xue-Na Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
- Jinling Pharmaceutical Co., Ltd., Nanjing, China
| |
Collapse
|
3
|
Chen J, Li CG, Yang LX, Qian Y, Zhu LW, Liu PY, Cao X, Wang Y, Zhu MS, Xu Y. MYPT1 SMKO Mice Function as a Novel Spontaneous Age- and Hypertension-Dependent Animal Model of CSVD. Transl Stroke Res 2024; 15:606-619. [PMID: 36843141 DOI: 10.1007/s12975-023-01142-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 02/14/2023] [Accepted: 02/17/2023] [Indexed: 02/28/2023]
Abstract
Cerebral small vessel disease (CSVD) is the most common progressive vascular disease that causes vascular dementia. Aging and hypertension are major contributors to CSVD, but the pathophysiological mechanism remains unclear, mainly due to the lack of an ideal animal model. Our previous study revealed that vascular smooth muscle cell (VSMC)-specific myosin phosphatase target subunit 1 (MYPT1) knockout (MYPT1SMKO) leads to constant hypertension, prompting us to explore whether hypertensive MYPT1SMKO mice can be considered a novel CSVD animal model. Here, we found that MYPT1SMKO mice displayed age-dependent CSVD-like neurobehaviors, including decreased motion speed, anxiety, and cognitive decline. MYPT1SMKO mice exhibited remarkable white matter injury compared with control mice, as shown by the more prominent loss of myelin at 12 months of age. Additionally, MYPT1SMKO mice were found to exhibit CSVD-like small vessel impairment, including intravascular hyalinization, perivascular space enlargement, and microbleed and blood-brain barrier (BBB) disruption. Last, our results revealed that the brain of MYPT1SMKO mice was characterized by an exacerbated inflammatory microenvironment, which is similar to patients with CSVD. In light of the above structural and functional phenotypes that closely mimic the conditions of human CSVD, we suggest that MYPT1SMKO mice are a novel age- and hypertension-dependent animal model of CSVD.
Collapse
Affiliation(s)
- Jian Chen
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Cheng-Gang Li
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Li-Xuan Yang
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Yi Qian
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Li-Wen Zhu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Pin-Yi Liu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Xiang Cao
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Ye Wang
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Min-Sheng Zhu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China.
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, China.
| | - Yun Xu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China.
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, China.
- Jiangsu Provincial Key Discipline of Neurology, Nanjing, 210008, China.
| |
Collapse
|
4
|
Lee E, May H, Kazmierczak K, Liang J, Nguyen N, Hill JA, Gillette TG, Szczesna-Cordary D, Chang AN. The MYPT2-regulated striated muscle-specific myosin light chain phosphatase limits cardiac myosin phosphorylation in vivo. J Biol Chem 2024; 300:105652. [PMID: 38224947 PMCID: PMC10851227 DOI: 10.1016/j.jbc.2024.105652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 01/02/2024] [Accepted: 01/04/2024] [Indexed: 01/17/2024] Open
Abstract
The physiological importance of cardiac myosin regulatory light chain (RLC) phosphorylation by its dedicated cardiac myosin light chain kinase has been established in both humans and mice. Constitutive RLC-phosphorylation, regulated by the balanced activities of cardiac myosin light chain kinase and myosin light chain phosphatase (MLCP), is fundamental to the biochemical and physiological properties of myofilaments. However, limited information is available on cardiac MLCP. In this study, we hypothesized that the striated muscle-specific MLCP regulatory subunit, MYPT2, targets the phosphatase catalytic subunit to cardiac myosin, contributing to the maintenance of cardiac function in vivo through the regulation of RLC-phosphorylation. To test this hypothesis, we generated a floxed-PPP1R12B mouse model crossed with a cardiac-specific Mer-Cre-Mer to conditionally ablate MYPT2 in adult cardiomyocytes. Immunofluorescence microscopy using the gene-ablated tissue as a control confirmed the localization of MYPT2 to regions where it overlaps with a subset of RLC. Biochemical analysis revealed an increase in RLC-phosphorylation in vivo. The loss of MYPT2 demonstrated significant protection against pressure overload-induced hypertrophy, as evidenced by heart weight, qPCR of hypertrophy-associated genes, measurements of myocyte diameters, and expression of β-MHC protein. Furthermore, mantATP chase assays revealed an increased ratio of myosin heads distributed to the interfilament space in MYPT2-ablated heart muscle fibers, confirming that RLC-phosphorylation regulated by MLCP, enhances cardiac performance in vivo. Our findings establish MYPT2 as the regulatory subunit of cardiac MLCP, distinct from the ubiquitously expressed canonical smooth muscle MLCP. Targeting MYPT2 to increase cardiac RLC-phosphorylation in vivo may improve baseline cardiac performance, thereby attenuating pathological hypertrophy.
Collapse
Affiliation(s)
- Eunyoung Lee
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Herman May
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Katarzyna Kazmierczak
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Jingsheng Liang
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Nhu Nguyen
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Joseph A Hill
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Thomas G Gillette
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Danuta Szczesna-Cordary
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Audrey N Chang
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Pak Center for Mineral Metabolism and Clinical Research, UTSW Medical Center, Dallas, Texas, USA.
| |
Collapse
|
5
|
Lee HH, Carmichael DJ, Ríbeiro V, Parisi DN, Munzen ME, Charles-Niño CL, Hamed MF, Kaur E, Mishra A, Patel J, Rooklin RB, Sher A, Carrillo-Sepulveda MA, Eugenin EA, Dores MR, Martinez LR. Glucuronoxylomannan intranasal challenge prior to Cryptococcus neoformans pulmonary infection enhances cerebral cryptococcosis in rodents. PLoS Pathog 2023; 19:e1010941. [PMID: 37115795 PMCID: PMC10171644 DOI: 10.1371/journal.ppat.1010941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 05/10/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
The encapsulated fungus Cryptococcus neoformans is the most common cause of fungal meningitis, with the highest rate of disease in patients with AIDS or immunosuppression. This microbe enters the human body via inhalation of infectious particles. C. neoformans capsular polysaccharide, in which the major component is glucuronoxylomannan (GXM), extensively accumulates in tissues and compromises host immune responses. C. neoformans travels from the lungs to the bloodstream and crosses to the brain via transcytosis, paracytosis, or inside of phagocytes using a "Trojan horse" mechanism. The fungus causes life-threatening meningoencephalitis with high mortality rates. Hence, we investigated the impact of intranasal exogenous GXM administration on C. neoformans infection in C57BL/6 mice. GXM enhances cryptococcal pulmonary infection and facilitates fungal systemic dissemination and brain invasion. Pre-challenge of GXM results in detection of the polysaccharide in lungs, serum, and surprisingly brain, the latter likely reached through the nasal cavity. GXM significantly alters endothelial cell tight junction protein expression in vivo, suggesting significant implications for the C. neoformans mechanisms of brain invasion. Using a microtiter transwell system, we showed that GXM disrupts the trans-endothelial electrical resistance, weakening human brain endothelial cell monolayers co-cultured with pericytes, supportive cells of blood vessels/capillaries found in the blood-brain barrier (BBB) to promote C. neoformans BBB penetration. Our findings should be considered in the development of therapeutics to combat the devastating complications of cryptococcosis that results in an estimated ~200,000 deaths worldwide each year.
Collapse
Affiliation(s)
- Hiu Ham Lee
- Department of Biomedical Sciences, NYIT College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, New York, United States of America
| | - Dylan J Carmichael
- Department of Biomedical Sciences, NYIT College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, New York, United States of America
| | - Victoria Ríbeiro
- Department of Neuroscience, Cell Biology, and Anatomy, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Dana N Parisi
- Department of Biomedical Sciences, NYIT College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, New York, United States of America
- Department of Biomedical Sciences, Long Island University-Post, Brookville, New York, United States of America
| | - Melissa E Munzen
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, United States of America
| | - Claudia L Charles-Niño
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, United States of America
| | - Mohamed F Hamed
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, United States of America
- Department of Pathology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Ettiman Kaur
- Department of Biology, Hofstra University, Hempstead, New York, United States of America
| | - Ayush Mishra
- Department of Biology, Hofstra University, Hempstead, New York, United States of America
| | - Jiya Patel
- Department of Biology, Hofstra University, Hempstead, New York, United States of America
| | - Rikki B Rooklin
- Department of Biology, Hofstra University, Hempstead, New York, United States of America
| | - Amina Sher
- Department of Biology, Hofstra University, Hempstead, New York, United States of America
| | - Maria A Carrillo-Sepulveda
- Department of Biomedical Sciences, NYIT College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, New York, United States of America
| | - Eliseo A Eugenin
- Department of Neuroscience, Cell Biology, and Anatomy, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Michael R Dores
- Department of Biology, Hofstra University, Hempstead, New York, United States of America
| | - Luis R Martinez
- Department of Biomedical Sciences, NYIT College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, New York, United States of America
- Department of Biomedical Sciences, Long Island University-Post, Brookville, New York, United States of America
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, United States of America
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, United States of America
- Center for Immunology and Transplantation, University of Florida, Gainesville, Florida, United States of America
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, Florida, United States of America
| |
Collapse
|
6
|
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
|
7
|
Hong SH, Jang EB, Hwang HJ, Park SY, Moon HS, Yoon YE. Effect of α1D-adrenoceptor blocker for the reduction of ureteral contractions. Investig Clin Urol 2023; 64:82-90. [PMID: 36629069 PMCID: PMC9834562 DOI: 10.4111/icu.20220254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/01/2022] [Accepted: 11/21/2022] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Urolithiasis is a common urinary tract disease with growing prevalence. Alpha1-adrenoceptors (α1-ARs) are abundant in ureteral smooth muscle, distributed with different α1-AR subtypes. α1D-AR is the most widely distributed in the ureter. However, the effect of α1D-AR blockade on ureteric contraction remains unknown. MATERIALS AND METHODS We dissected smooth muscle tissues (3 mm×3 mm) from the rat bladder and human ureter, tied silk strips on both tissue ends, and measured contraction in an organ bath chamber. Contraction activity in ureteral smooth muscle cells (USMCs) was immunocytochemically examined using primary rat and human USMC cultures. RESULTS Using the organ bath system, we determined the inhibitory effects of silodosin, tamsulosin, and naftopidil. Naftopidil significantly decreased contractility of rat bladder tissue; similar results were observed in human ureteral tissue. To confirm ex vivo experimental results in vitro , we examined the phosphorylation of myosin light chain (MLC), a marker of contractility, in a primary human USMC culture. The examined drugs decreased phospho-MLC levels in human USMCs; however, naftopidil profoundly increased MLC dephosphorylation. CONCLUSIONS We studied the effects of naftopidil, an α1D-AR inhibitor, on the ureter. Compared with alpha-blockers, naftopidil significantly relaxed ureteral smooth muscle. Therefore, naftopidil could be an effective therapy for patients with ureteral stones.
Collapse
Affiliation(s)
- Seong Hwi Hong
- Department of Urology, Hanyang University College of Medicine, Seoul, Korea
| | - Eun Bi Jang
- Department of Urology, Hanyang University College of Medicine, Seoul, Korea.,Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul, Korea
| | - Hyun Ji Hwang
- Department of Urology, Hanyang University College of Medicine, Seoul, Korea.,Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul, Korea
| | - Sung Yul Park
- Department of Urology, Hanyang University College of Medicine, Seoul, Korea
| | - Hong Sang Moon
- Department of Urology, Hanyang University College of Medicine, Seoul, Korea
| | - Young Eun Yoon
- Department of Urology, Hanyang University College of Medicine, Seoul, Korea.,Department of Medical and Digital Engineering, Hanyang University Graduate School, Seoul, Korea
| |
Collapse
|
8
|
Zhao W, Sun J, Yao LY, Hang D, Li YQ, Chen CP, Zhou YW, Chen X, Tao T, Wei LS, Zheng YY, Ge X, Li CJ, Xin ZC, Pan Y, Wang XZ, He WQ, Zhang XN, Yao B, Zhu MS. MYPT1 reduction is a pathogenic factor of erectile dysfunction. Commun Biol 2022; 5:744. [PMID: 35879418 PMCID: PMC9314386 DOI: 10.1038/s42003-022-03716-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 07/14/2022] [Indexed: 11/09/2022] Open
Abstract
Erectile dysfunction (ED) is closely associated with smooth muscle dysfunction, but its underlying mechanisms remains incompletely understood. We here reported that the reduced expression of myosin phosphatase target subunit 1 (MYPT1), the main regulatory unit of myosin light chain phosphatase, was critical for the development of vasculogenic ED. Male MYPT1 knockout mice had reduced fertility and the penises displayed impaired erections as evidenced by reduced intracavernous pressure (ICP). The penile smooth muscles of the knockout mice displayed enhanced response to G-Protein Couple Receptor agonism and depolarization contractility and resistant relaxation. We further identified a natural compound lotusine that increased the MYPT1 expression by inhibiting SIAH1/2 E3 ligases-mediated protein degradation. This compound sufficiently restored the ICP and improved histological characters of the penile artery of Mypt1 haploinsufficiency mice. In diabetic ED mice (db/db), the decreased expression of MYPT1 was measured, and ICP was improved by lotusine treatment. We conclude that the reduction of MYPT1 is the major pathogenic factor of vasculogenic ED. The restoration of MYPT1 by lotusine improved the function of injured penile smooth muscles, and could be a novel strategy for ED therapy.
Collapse
Affiliation(s)
- Wei Zhao
- Jinling Hospital Department of Reproductive Medical Center affiliated Sch Med, State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Jie Sun
- Jinling Hospital Department of Reproductive Medical Center affiliated Sch Med, State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Liang-Yu Yao
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Dong Hang
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Ye-Qiong Li
- Jinling Hospital Department of Reproductive Medical Center affiliated Sch Med, State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Cai-Ping Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Yu-Wei Zhou
- Jinling Hospital Department of Reproductive Medical Center affiliated Sch Med, State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Xin Chen
- Jinling Hospital Department of Reproductive Medical Center affiliated Sch Med, State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Tao Tao
- Jinling Hospital Department of Reproductive Medical Center affiliated Sch Med, State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Li-Sha Wei
- Jinling Hospital Department of Reproductive Medical Center affiliated Sch Med, State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Yan-Yan Zheng
- Jinling Hospital Department of Reproductive Medical Center affiliated Sch Med, State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Xie Ge
- Jinling Hospital Department of Reproductive Medical Center affiliated Sch Med, State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Chao-Jun Li
- Jinling Hospital Department of Reproductive Medical Center affiliated Sch Med, State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Zhong-Cheng Xin
- Andrology Center, Peking University First Hospital, Peking University, Beijing, China
| | - Yang Pan
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xin-Zhu Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wei-Qi He
- Cambridge-Suda (CAM-SU) Genomic Resource Center, Soochow University, Suzhou, China
| | - Xue-Na Zhang
- Jinling Hospital Department of Reproductive Medical Center affiliated Sch Med, State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China.
| | - Bing Yao
- Jinling Hospital Department of Reproductive Medical Center affiliated Sch Med, State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China.
| | - Min-Sheng Zhu
- Jinling Hospital Department of Reproductive Medical Center affiliated Sch Med, State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China.
| |
Collapse
|
9
|
Sani MB, Roudbari Z, Karimi O, Banabazi MH, Esmaeilkhanian S, Asadzadeh N, Zare Harofte J, Shafei Naderi A, Burger PA. Gene-Set Enrichment Analysis for Identifying Genes and Biological Activities Associated with Growth Traits in Dromedaries. Animals (Basel) 2022; 12:184. [PMID: 35049806 PMCID: PMC8773174 DOI: 10.3390/ani12020184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/09/2022] [Accepted: 01/11/2022] [Indexed: 11/16/2022] Open
Abstract
Growth is an important heritable economic trait for dromedaries and necessary for planning a successful breeding program. Until now, genome-wide association studies (GWAS) and QTL-mapping have identified significant single nucleotide polymorphisms (SNPs) associated with growth in domestic animals, but in dromedaries, the number of studies is very low. This project aimed to find biological themes affecting growth in dromedaries. In the first step, 99 candidate SNPs were chosen from a previously established set of SNPs associated with body weight, gain, and birth weight in Iranian dromedaries. Next, 0.5 kb upstream and downstream of each candidate SNP were selected from NCBI (assembly accession: GCA_000803125.3). The annotation of fragments with candidate SNPs regarding the reference genome was retrieved using the Blast2GO tool. Candidate SNPs associated with growth were mapped to 22 genes, and 25 significant biological themes were identified to be related to growth in dromedaries. The main biological functions included calcium ion binding, protein binding, DNA-binding transcription factor activity, protein kinase activity, tropomyosin binding, myosin complex, actin-binding, ATP binding, receptor signaling pathway via JAK-STAT, and cytokine activity. EFCAB5, MTIF2, MYO3A, TBX15, IFNL3, PREX1, and TMOD3 genes are candidates for improving growth in camel breeding programs.
Collapse
Affiliation(s)
- Morteza Bitaraf Sani
- Animal Science Research Department, Yazd Agricultural and Natural Resources Research and Education Center, Agricultural Research, Education & Extension Organization (AREEO), Yazd 8915813155, Iran; (J.Z.H.); (A.S.N.)
| | - Zahra Roudbari
- Department of Animal Science, Faculty of Agriculture, University of Jiroft, Jiroft 7867155311, Iran;
| | - Omid Karimi
- Department of Animal Viral Diseases Research, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization, Karaj 3146618361, Iran;
| | - Mohammad Hossein Banabazi
- Animal Science Research Institute of Iran, Agricultural Research, Education and Extension Organization (AREEO), Karaj 3146618361, Iran; (M.H.B.); (S.E.); (N.A.)
- Department of Animal Breeding and Genetics (HGEN), Centre for Veterinary Medicine and Animal Science (VHC), Swedish University of Agricultural Sciences (SLU), 75007 Uppsala, Sweden
| | - Saeid Esmaeilkhanian
- Animal Science Research Institute of Iran, Agricultural Research, Education and Extension Organization (AREEO), Karaj 3146618361, Iran; (M.H.B.); (S.E.); (N.A.)
| | - Nader Asadzadeh
- Animal Science Research Institute of Iran, Agricultural Research, Education and Extension Organization (AREEO), Karaj 3146618361, Iran; (M.H.B.); (S.E.); (N.A.)
| | - Javad Zare Harofte
- Animal Science Research Department, Yazd Agricultural and Natural Resources Research and Education Center, Agricultural Research, Education & Extension Organization (AREEO), Yazd 8915813155, Iran; (J.Z.H.); (A.S.N.)
| | - Ali Shafei Naderi
- Animal Science Research Department, Yazd Agricultural and Natural Resources Research and Education Center, Agricultural Research, Education & Extension Organization (AREEO), Yazd 8915813155, Iran; (J.Z.H.); (A.S.N.)
| | - Pamela Anna Burger
- Research Institute of Wildlife Ecology, Vetmeduni Vienna, 1160 Vienna, Austria;
| |
Collapse
|
10
|
Regulation of myosin light-chain phosphorylation and its roles in cardiovascular physiology and pathophysiology. Hypertens Res 2022; 45:40-52. [PMID: 34616031 DOI: 10.1038/s41440-021-00733-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 06/19/2021] [Accepted: 07/08/2021] [Indexed: 01/22/2023]
Abstract
The regulation of muscle contraction is a critical function in the cardiovascular system, and abnormalities may be life-threatening or cause illness. The common basic mechanism in muscle contraction is the interaction between the protein filaments myosin and actin. Although this interaction is primarily regulated by intracellular Ca2+, the primary targets and intracellular signaling pathways differ in vascular smooth muscle and cardiac muscle. Phosphorylation of the myosin regulatory light chain (RLC) is a primary molecular switch for smooth muscle contraction. The equilibrium between phosphorylated and unphosphorylated RLC is dynamically achieved through two enzymes, myosin light chain kinase, a Ca2+-dependent enzyme, and myosin phosphatase, which modifies the Ca2+ sensitivity of contractions. In cardiac muscle, the primary target protein for Ca2+ is troponin C on thin filaments; however, RLC phosphorylation also plays a modulatory role in contraction. This review summarizes recent advances in our understanding of the regulation, physiological function, and pathophysiological involvement of RLC phosphorylation in smooth and cardiac muscles.
Collapse
|
11
|
Lubomirov LT, Jänsch MH, Papadopoulos S, Schroeter MM, Metzler D, Bust M, Hescheler J, Grisk O, Ritter O, Pfitzer G. Senescent murine femoral arteries undergo vascular remodelling associated with accelerated stress-induced contractility and reactivity to nitric oxide. Basic Clin Pharmacol Toxicol 2021; 130:70-83. [PMID: 34665520 DOI: 10.1111/bcpt.13675] [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: 05/27/2021] [Indexed: 12/17/2022]
Abstract
This work explored the mechanism of augmented stress-induced vascular reactivity of senescent murine femoral arteries (FAs). Mechanical and pharmacological reactivity of young (12-25 weeks, y-FA) and senescent (>104 weeks, s-FAs) femoral arteries was measured by wire myography. Expression and protein phosphorylation of selected regulatory proteins were studied by western blotting. Expression ratio of the Exon24 in/out splice isoforms of the regulatory subunit of myosin phosphatase, MYPT1 (MYPT1-Exon24 in/out), was determined by polymerase chain reaction (PCR). While the resting length-tension relationship showed no alteration, the stretch-induced-tone increased to 8.3 ± 0.9 mN in s-FA versus only 4.6 ± 0.3 mN in y-FAs. Under basal conditions, phosphorylation of the regulatory light chain of myosin at S19 was 19.2 ± 5.8% in y-FA versus 49.2 ± 12.6% in s-FA. Inhibition of endogenous NO release raised tone additionally to 10.4 ± 1.2 mN in s-FA, whereas this treatment had a negligible effect in y-FAs (4.8 ± 0.3 mN). In s-FAs, reactivity to NO donor was augmented (pD2 = -4.5 ± 0.3 in y-FA vs. -5.2 ± 0.1 in senescent). Accordingly, in s-FAs, MYPT1-Exon24-out-mRNA, which is responsible for expression of the more sensitive to protein-kinase G, leucine-zipper-positive MYPT1 isoform, was increased. The present work provides evidence that senescent murine s-FA undergoes vascular remodelling associated with increases in stretch-activated contractility and sensitivity to NO/cGMP/PKG system.
Collapse
Affiliation(s)
- Lubomir T Lubomirov
- Institute of Physiology, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany.,Institute of Vegetative Physiology, Center of Physiology, University of Cologne, Cologne, Germany.,Research Cluster, Molecular Mechanisms of Cardiovascular Diseases, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
| | - Monique Heidrun Jänsch
- Research Cluster, Molecular Mechanisms of Cardiovascular Diseases, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany.,Department of Cardiology, Nephrology and Pneumology, Brandenburg Medical School, University Hospital Brandenburg, Brandenburg an der Havel, Germany
| | - Symeon Papadopoulos
- Institute of Neurophysiology, Center of Physiology, University of Cologne, Cologne, Germany
| | - Mechthild M Schroeter
- Institute of Vegetative Physiology, Center of Physiology, University of Cologne, Cologne, Germany
| | - Doris Metzler
- Institute of Vegetative Physiology, Center of Physiology, University of Cologne, Cologne, Germany
| | - Maria Bust
- Institute of Vegetative Physiology, Center of Physiology, University of Cologne, Cologne, Germany
| | - Jürgen Hescheler
- Institute of Neurophysiology, Center of Physiology, 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
| | - Oliver Ritter
- Research Cluster, Molecular Mechanisms of Cardiovascular Diseases, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany.,Department of Cardiology, Nephrology and Pneumology, Brandenburg Medical School, University Hospital Brandenburg, Brandenburg an der Havel, Germany
| | - Gabriele Pfitzer
- Institute of Vegetative Physiology, Center of Physiology, University of Cologne, Cologne, Germany
| |
Collapse
|
12
|
Selivanova EK, Gaynullina DK, Tarasova OS. Thyroxine Induces Acute Relaxation of Rat Skeletal Muscle Arteries via Integrin αvβ3, ERK1/2 and Integrin-Linked Kinase. Front Physiol 2021; 12:726354. [PMID: 34594239 PMCID: PMC8477044 DOI: 10.3389/fphys.2021.726354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/23/2021] [Indexed: 11/17/2022] Open
Abstract
Aim: Hyperthyroidism is associated with a decreased peripheral vascular resistance, which could be caused by the vasodilator genomic or non-genomic effects of thyroid hormones (TH). Non-genomic, or acute, effects develop within several minutes and involve a wide tissue-specific spectrum of molecular pathways poorly studied in vasculature. We aimed to investigate the mechanisms of acute effects of TH on rat skeletal muscle arteries. Methods: Sural arteries from male Wistar rats were used for isometric force recording (wire myography) and phosphorylated protein content measurement (Western blotting). Results: Both triiodothyronine (T3) and thyroxine (T4) reduced contractile response of sural arteries to α1-adrenoceptor agonist methoxamine. The effect of T4 was more prominent than T3 and not affected by iopanoic acid, an inhibitor of deiodinase 2. Endothelium denudation abolished the effect of T3, but not T4. Integrin αvβ3 inhibitor tetrac abolished the effect of T4 in endothelium-denuded arteries. T4 weakened methoxamine-induced elevation of phospho-MLC2 (Ser19) content in arterial samples. The effect of T4 in endothelium-denuded arteries was abolished by inhibiting ERK1/2 activation with U0126 as well as by ILK inhibitor Cpd22 but persisted in the presence of Src- or Rho-kinase inhibitors (PP2 and Y27632, respectively). Conclusion: Acute non-genomic relaxation of sural arteries induced by T3 is endothelium-dependent and that induced by T4 is endothelium-independent. The effect of T4 on α1-adrenergic contraction is stronger compared to T3 and involves the suppression of extracellular matrix signaling via integrin αvβ3, ERK1/2 and ILK with subsequent decrease of MLC2 (Ser19) phosphorylation.
Collapse
Affiliation(s)
- Ekaterina K Selivanova
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Dina K Gaynullina
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia.,Department of Physiology, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Olga S Tarasova
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia.,Laboratory of Exercise Physiology, Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
13
|
Pütz S, Barthel LS, Frohn M, Metzler D, Barham M, Pryymachuk G, Trunschke O, Lubomirov LT, Hescheler J, Chalovich JM, Neiss WF, Koch M, Schroeter MM, Pfitzer G. Caldesmon ablation in mice causes umbilical herniation and alters contractility of fetal urinary bladder smooth muscle. J Gen Physiol 2021; 153:212279. [PMID: 34115104 PMCID: PMC8203487 DOI: 10.1085/jgp.202012776] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 04/21/2021] [Indexed: 12/13/2022] Open
Abstract
The actin-, myosin-, and calmodulin-binding protein caldesmon (CaD) is expressed in two splice isoforms: h-CaD, which is an integral part of the actomyosin domain of smooth muscle cells, and l-CaD, which is widely expressed and is involved in many cellular functions. Despite extensive research for many years, CaD's in vivo function has remained elusive. To explore the role of CaD in smooth muscle contraction in vivo, we generated a mutant allele that ablates both isoforms. Heterozygous animals were viable and had a normal life span, but homozygous mutants died perinatally, likely because of a persistent umbilical hernia. The herniation was associated with hypoplastic and dysmorphic abdominal wall muscles. We assessed mechanical parameters in isometrically mounted longitudinal strips of E18.5 urinary bladders and in ring preparations from abdominal aorta using wire myography. Ca2+ sensitivity was higher and relaxation rate was slower in Cald1−/− compared with Cald1+/+ skinned bladder strips. However, we observed no change in the content and phosphorylation of regulatory proteins of the contractile apparatus and myosin isoforms known to affect these contractile parameters. Intact fibers showed no difference in actin and myosin content, regardless of genotype, although KCl-induced force tended to be lower in homozygous and higher in heterozygous mutants than in WTs. Conversely, in skinned fibers, myosin content and maximal force were significantly lower in Cald1−/− than in WTs. In KO abdominal aortas, resting and U46619 elicited force were lower than in WTs. Our results are consistent with the notion that CaD impacts smooth muscle function dually by (1) acting as a molecular brake on contraction and (2) maintaining the structural integrity of the contractile machinery. Most importantly, CaD is essential for resolution of the physiological umbilical hernia and ventral body wall closure.
Collapse
Affiliation(s)
- Sandra Pütz
- Institute of Vegetative Physiology, Center of Physiology, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Lisa Sophie Barthel
- Institute of Vegetative Physiology, Center of Physiology, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Marina Frohn
- Institute of Vegetative Physiology, Center of Physiology, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Doris Metzler
- Institute of Vegetative Physiology, Center of Physiology, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Mohammed Barham
- Institute of Anatomy I, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Galyna Pryymachuk
- Institute of Anatomy I, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Oliver Trunschke
- Institute of Vegetative Physiology, Center of Physiology, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Lubomir T Lubomirov
- Institute of Vegetative Physiology, Center of Physiology, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Jürgen Hescheler
- Institute of Neurophysiology, Center of Physiology, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Joseph M Chalovich
- Department of Biochemistry & Molecular Biology, Brody School of Medicine at East Carolina University, Greenville, NC
| | - Wolfram F Neiss
- Institute of Anatomy I, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Manuel Koch
- Institute for Dental Research and Oral Musculoskeletal Biology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Mechthild M Schroeter
- Institute of Vegetative Physiology, Center of Physiology, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Gabriele Pfitzer
- Institute of Vegetative Physiology, Center of Physiology, Faculty of Medicine, University of Cologne, Cologne, Germany
| |
Collapse
|
14
|
LIMK2 is required for membrane cytoskeleton reorganization of contracting airway smooth muscle. J Genet Genomics 2021; 48:452-462. [PMID: 34353741 DOI: 10.1016/j.jgg.2021.04.014] [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: 12/14/2020] [Revised: 04/14/2021] [Accepted: 04/26/2021] [Indexed: 11/23/2022]
Abstract
Airway smooth muscle (ASM) has developed a mechanical adaption mechanism by which it transduces force and responds to environmental forces, which is essential for periodic breathing. Cytoskeletal reorganization has been implicated in this process, but the regulatory mechanism remains to be determined. We here observe that ASM abundantly expresses cytoskeleton regulators Limk1 and Limk2, and their expression levels are further upregulated in chronic obstructive pulmonary disease (COPD) animals. By establishing mouse lines with deletions of Limk1 or Limk2, we analyse the length-sensitive contraction, F/G-actin dynamics, and F-actin pool of mutant ASM cells. As LIMK1 phosphorylation does not respond to the contractile stimulation, LIMK1-deficient ASM develops normal maximal force, while LIMK2 or LIMK1/LIMK2 deficient ASMs show approximately 30% inhibition. LIMK2 deletion causes a significant decrease in cofilin phosphorylation along with a reduced F/G-actin ratio. As LIMK2 functions independently of cross-bridge movement, this observation indicates that LIMK2 is necessary for F-actin dynamics and hence force transduction. Moreover, LIMK2-deficient ASMs display abolishes stretching-induced suppression of 5-hydroxytryptamine (5-HT) but not acetylcholine-evoks force, which is due to the differential contraction mechanisms adopted by the agonists. We propose that LIMK2-mediated cofilin phosphorylation is required for membrane cytoskeleton reorganization that is necessary for ASM mechanical adaption including the 5-HT-evoked length-sensitive effect.
Collapse
|
15
|
Mfge8 attenuates human gastric antrum smooth muscle contractions. J Muscle Res Cell Motil 2021; 42:219-231. [PMID: 34085177 PMCID: PMC8332633 DOI: 10.1007/s10974-021-09604-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 05/21/2021] [Indexed: 12/02/2022]
Abstract
Coordinated gastric smooth muscle contraction is critical for proper digestion and is adversely affected by a number of gastric motility disorders. In this study we report that the secreted protein Mfge8 (milk fat globule-EGF factor 8) inhibits the contractile responses of human gastric antrum muscles to cholinergic stimuli by reducing the inhibitory phosphorylation of the MYPT1 (myosin phosphatase-targeting subunit (1) subunit of MLCP (myosin light chain phosphatase), resulting in reduced LC20 (smooth muscle myosin regulatory light chain (2) phosphorylation. Mfge8 reduced the agonist-induced increase in the F-actin/G-actin ratios of β-actin and γ-actin1. We show that endogenous Mfge8 is bound to its receptor, α8β1 integrin, in human gastric antrum muscles, suggesting that human gastric antrum muscle mechanical responses are regulated by Mfge8. The regulation of gastric antrum smooth muscles by Mfge8 and α8 integrin functions as a brake on gastric antrum mechanical activities. Further studies of the role of Mfge8 and α8 integrin in regulating gastric antrum function will likely reveal additional novel aspects of gastric smooth muscle motility mechanisms.
Collapse
|
16
|
Wei X, Lan T, Zhou Y, Cheng J, Li P, Zeng X, Yang Y. Mechanism of α1-Adrenergic Receptor-Induced Increased Contraction of Rat Mesenteric Artery in Aging Hypertension Rats. Gerontology 2021; 67:323-337. [PMID: 33752204 DOI: 10.1159/000511911] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 09/17/2020] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Vasoconstriction is triggered by an increase in intracellular-free calcium concentration. Growing evidence indicates that contraction is also regulated by calcium-independent mechanisms involving RhoA-Rho kinase (ROCK), protein kinase C (PKC), and so on. In this study, we studied the changes of vascular reactivity as well as the underlying signaling pathways in aging spontaneously hypertensive rats (SHRs). METHODS The artery tension induced by α1-adrenergic receptor activator (α1-AR) phenylephrine (PE) was measured in the absence or presence of myosin light chain kinase (MLCK), PKC, and ROCK inhibitors. The α1-AR, PKC, ROCK, phosphorylation of myosin light chain (MLC), and PKC-potentiated phosphatase inhibitors of 17 kDa (CPI-17) of rat mesenteric arteries were analyzed at the mRNA level or protein level. RESULTS The vascular tension measurements showed that there was a significant increase in the mesenteric artery contraction induced by PE in old SHR. MLCK inhibitor ML-7 can similarly inhibit PE-induced vasoconstriction. PKC inhibitor GF109203X has the weakest inhibitory effect on PE-induced contraction in old SHR. At the presence of ROCK inhibitor H1152, PE-induced contraction was significantly reduced in young Wistar-Kyoto (WKY) rats, but this phenomenon disappeared in other rats. Furthermore, in old SHR the protein expression of α1-AR decreased and phosphorylation of MLC and CPI-17 were upregulated and MLC phosphatase (MLCP) activity was significantly lower. The expressions of PKC were upregulated in SHR and old rats. In addition, the expression of ROCK-1 was decreased and ROCK-2 was significantly upregulated with age in SHR. CONCLUSION In aging hypertension, the expression/activity of PKC or ROCK-2/CPI-17 excessively increased, MLCP activity decreased and MLC phosphorylation enhanced, leading to increased α1-AR-induced vasoconstriction.
Collapse
Affiliation(s)
- Xiaoyu Wei
- Key Lab 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, Luzhou, China
| | - Ting Lan
- Key Lab 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, Luzhou, China
| | - Yuanqun Zhou
- Key Lab 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, Luzhou, China
| | - Jun Cheng
- Key Lab 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, Luzhou, China
| | - Pengyun Li
- Key Lab 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, Luzhou, China
| | - Xiaorong Zeng
- Key Lab 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, Luzhou, China
| | - Yan Yang
- Key Lab 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, Luzhou, China,
| |
Collapse
|
17
|
Sun J, Qiao YN, Tao T, Zhao W, Wei LS, Li YQ, Wang W, Wang Y, Zhou YW, Zheng YY, Chen X, Pan HC, Zhang XN, Zhu MS. Distinct Roles of Smooth Muscle and Non-muscle Myosin Light Chain-Mediated Smooth Muscle Contraction. Front Physiol 2020; 11:593966. [PMID: 33424621 PMCID: PMC7793928 DOI: 10.3389/fphys.2020.593966] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/18/2020] [Indexed: 12/17/2022] Open
Abstract
Both smooth muscle (SM) and non-muscle (NM) myosin II are expressed in hollow organs such as the bladder and uterus, but their respective roles in contraction and corresponding physiological functions remain to be determined. In this report, we assessed their roles by analyzing mice deficient of Myl9, a gene encoding the SM myosin regulatory light chain (SM RLC). We find that global Myl9-deficient bladders contracted with an apparent sustained phase, despite no initial phase. This sustained contraction was mediated by NM myosin RLC (NM RLC) phosphorylation by myosin light chain kinase (MLCK). NM myosin II was expressed abundantly in the uterus and young mice bladders, of which the force was accordingly sensitive to NM myosin inhibition. Our findings reveal distinct roles of SM RLC and NM RLC in SM contraction.
Collapse
Affiliation(s)
- Jie Sun
- Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Yan-Ning Qiao
- Key Laboratory of MOE for Modern Teaching Technology, Shaanxi Normal University, Xi'an, China
| | - Tao Tao
- Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Wei Zhao
- Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Li-Sha Wei
- Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Ye-Qiong Li
- Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Wei Wang
- Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Ye Wang
- Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Yu-Wei Zhou
- Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Yan-Yan Zheng
- Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Xin Chen
- Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Hong-Chun Pan
- College of Life Sciences, Anhui Normal University, Wuhu, China
| | - Xue-Na Zhang
- Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Min-Sheng Zhu
- Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| |
Collapse
|
18
|
Álvarez-Santos MD, Álvarez-González M, Estrada-Soto S, Bazán-Perkins B. Regulation of Myosin Light-Chain Phosphatase Activity to Generate Airway Smooth Muscle Hypercontractility. Front Physiol 2020; 11:701. [PMID: 32676037 PMCID: PMC7333668 DOI: 10.3389/fphys.2020.00701] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 05/28/2020] [Indexed: 12/21/2022] Open
Abstract
Smooth muscle is a central structure involved in the regulation of airway tone. In addition, it plays an important role in the development of some pathologies generated by alterations in contraction, such as hypercontractility and the airway hyperresponsiveness observed in asthma. The molecular processes associated with smooth muscle contraction are centered around myosin light chain (MLC) phosphorylation, which is controlled by a balance in the activity of myosin light-chain kinase (MLCK) and myosin light-chain phosphatase (MLCP). MLCK activation depends on increasing concentrations of intracellular Ca2+, while MLCP activation is independent of Ca2+. MLCP contains a phosphatase subunit (PP1c) that is regulated through myosin phosphatase target subunit 1 (MYPT1) and other subunits, such as glycogen-associated regulatory subunit and myosin-binding subunit 85 kDa. Interestingly, MLCP inhibition may contribute to exacerbation of smooth muscle contraction by increasing MLC phosphorylation to induce hypercontractility. Many pathways inhibiting MLCP activity in airway smooth muscle have been proposed and are focused on inhibition of PP1c, inhibitory phosphorylation of MYPT1 and dissociation of the PP1c-MYPT1 complex.
Collapse
Affiliation(s)
- Mayra D Álvarez-Santos
- Biology Area, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Marisol Álvarez-González
- Laboratorio de Inmunofarmacología, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Mexico City, Mexico
| | - Samuel Estrada-Soto
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Mexico
| | - Blanca Bazán-Perkins
- Laboratorio de Inmunofarmacología, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Mexico City, Mexico.,Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Mexico
| |
Collapse
|
19
|
Valdés A, Zhao H, Pettersson U, Lind SB. Phosphorylation Time-Course Study of the Response during Adenovirus Type 2 Infection. Proteomics 2020; 20:e1900327. [PMID: 32032466 DOI: 10.1002/pmic.201900327] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 01/22/2020] [Indexed: 12/31/2022]
Abstract
PTMs such as phosphorylations are usually involved in signal transduction pathways. To investigate the temporal dynamics of phosphoproteome changes upon viral infection, a model system of IMR-90 cells infected with human adenovirus type 2 (Ad2) is used in a time-course quantitative analysis combining titanium dioxide (TiO2 ) particle enrichment and SILAC-MS. Quantitative data from 1552 phosphorylated sites clustered the highly altered phosphorylated sites to the signaling by rho family GTPases, the actin cytoskeleton signaling, and the cAMP-dependent protein kinase A signaling pathways. Their activation is especially pronounced at early time post-infection. Changes of several phosphorylated sites involved in the glycolysis pathway, related to the activation of the Warburg effect, point at virus-induced energy production. For Ad2 proteins, 32 novel phosphorylation sites are identified and as many as 52 phosphorylated sites on 17 different Ad2 proteins are quantified, most of them at late time post-infection. Kinase predictions highlighted activation of PKA, CDK1/2, MAPK, and CKII. Overlaps of kinase motif sequences for viral and human proteins are observed, stressing the importance of phosphorylation during Ad2 infection.
Collapse
Affiliation(s)
- Alberto Valdés
- Section of Analytical Chemistry, Department of Chemistry-BMC, Uppsala University, Uppsala, 751 24, Sweden.,Department of Analytical Chemistry, Physical Chemistry and Chemical Engineering, University of Alcalá, Ctra. Madrid-Barcelona, Km. 33.600, 28871, Alcalá de Henares, Madrid, Spain
| | - Hongxing Zhao
- The Beijer Laboratory, Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Rudbeck Laboratory, 75185, Uppsala, Sweden
| | - Ulf Pettersson
- The Beijer Laboratory, Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Rudbeck Laboratory, 75185, Uppsala, Sweden
| | - Sara Bergström Lind
- Section of Analytical Chemistry, Department of Chemistry-BMC, Uppsala University, Uppsala, 751 24, Sweden
| |
Collapse
|
20
|
Lee CX, Cheah JH, Soule CK, Ding H, Whittaker CA, Karhohs K, Burds AA, Subramanyam KS, Carpenter AE, Eisner BH, Cima MJ. Identification and local delivery of vasodilators for the reduction of ureteral contractions. Nat Biomed Eng 2019; 4:28-39. [PMID: 31792422 DOI: 10.1038/s41551-019-0482-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 10/15/2019] [Indexed: 01/23/2023]
Abstract
Kidney stones and ureteral stents can cause ureteral colic and pain. By decreasing contractions in the ureter, clinically prescribed oral vasodilators may improve spontaneous stone passage rates and reduce the pain caused by ureteral stenting. We hypothesized that ureteral relaxation can be improved via the local administration of vasodilators and other smooth muscle relaxants. Here, by examining 18 candidate small molecules in an automated screening assay to determine the extent of ureteral relaxation, we show that the calcium channel blocker nifedipine and the Rho-kinase inhibitor ROCKi significantly relax human ureteral smooth muscle cells. We also show, by using ex vivo porcine ureter segments and sedated pigs that, with respect to the administration of a placebo, the local delivery of a clinically deployable formulation of the two drugs reduced ureteral contraction amplitude and frequency by 90% and 50%, respectively. Finally, we show that standard oral vasodilator therapy reduced contraction amplitude by only 50% and had a minimal effect on contraction frequency. Locally delivered ureteral relaxants therefore may improve ureter-related conditions.
Collapse
Affiliation(s)
- Christopher X Lee
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA.,David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jaime H Cheah
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Christian K Soule
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Huiming Ding
- The Barbara K. Ostrom (1978) Bioinformatics and Computing Facility in the Swanson Biotechnology Center, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Charles A Whittaker
- The Barbara K. Ostrom (1978) Bioinformatics and Computing Facility in the Swanson Biotechnology Center, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kyle Karhohs
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Aurora A Burds
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kriti S Subramanyam
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA.,David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Anne E Carpenter
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Brian H Eisner
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael J Cima
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA. .,David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA. .,Department of Material Science Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
21
|
Chen CP, Chen K, Feng Z, Wen X, Sun H. Synergistic antitumor activity of artesunate and HDAC inhibitors through elevating heme synthesis via synergistic upregulation of ALAS1 expression. Acta Pharm Sin B 2019; 9:937-951. [PMID: 31649844 PMCID: PMC6804493 DOI: 10.1016/j.apsb.2019.05.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 05/08/2019] [Accepted: 05/10/2019] [Indexed: 12/31/2022] Open
Abstract
Artemisinin and its derivatives (ARTs) were reported to display heme-dependent antitumor activity. On the other hand, histone deacetylase inhibitors (HDACi) were known to be able to promote heme synthesis in erythroid cells. Nevertheless, the effect of HDACi on heme homeostasis in non-erythrocytes remains unknown. We envisioned that the combination of HDACi and artesunate (ARS) might have synergistic antitumor activity through modulating heme synthesis. In vitro studies revealed that combination of ARS and HDACi exerted synergistic tumor inhibition by inducing cell death. Moreover, this combination exhibited more effective antitumor activity than either ARS or HDACi monotherapy in xenograft models without apparent toxicity. Importantly, mechanistic studies revealed that HDACi coordinated with ARS to increase 5-aminolevulinate synthase (ALAS1) expression, and subsequent heme production, leading to enhanced cytotoxicity of ARS. Notably, knocking down ALAS1 significantly blunted the synergistic effect of ARS and HDACi on tumor inhibition, indicating a critical role of ALAS1 upregulation in mediating ARS cytotoxicity. Collectively, our study revealed the mechanism of synergistic antitumor action of ARS and HDACi. This finding indicates that modulation of heme synthesis pathway by the combination based on ARTs and other heme synthesis modulators represents a promising therapeutic approach to solid tumors.
Collapse
Key Words
- ALA, 5-aminolevulinic acid
- ALAD, 5-aminolevulinate dehydratase
- ALAS, 5-aminolevulinate synthase
- ALAS1
- ARS, artesunate
- ART, artemisinin
- Antitumor
- Artesunate
- CCK-8, cell counting kit 8
- CI, combination index
- CMCNa, carboxymethyl cellulose
- DHA, dihydroartemisinin
- DMAB, (dimethylamino)benzaldehyde
- FECH, ferrochelatase
- GSDME, gasdermin E
- HDAC inhibitor
- HDAC, histone deacetylase
- HDACi, HDAC inhibitor
- HMBS, hydroxymethylbilane synthase
- Heme
- KD, knockdown
- KO, knockout
- LBH589, panobinostat
- PDT, photodynamic therapy
- PI, propidium iodide
- PpIX, protoporphyrin IX
- ROS, reactive oxygen species
- SA, succinyl acetone
- SAHA, vorinostat
- WT, wild-type
- sgRNA, single guide RNA
Collapse
Affiliation(s)
| | | | | | | | - Hongbin Sun
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
22
|
Li EQ, Zhao W, Zhang C, Qin LZ, Liu SJ, Feng ZQ, Wen X, Chen CP. Synthesis and anti-cancer activity of ND-646 and its derivatives as acetyl-CoA carboxylase 1 inhibitors. Eur J Pharm Sci 2019; 137:105010. [PMID: 31325544 DOI: 10.1016/j.ejps.2019.105010] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 07/09/2019] [Accepted: 07/15/2019] [Indexed: 12/31/2022]
Abstract
Acetyl-coA carboxylase 1 (ACC1) is the first and rate-limiting enzyme in the de novo fatty acid synthesis (FASyn) pathway. In this study, through public database analysis and clinic sample test, we for the first time verified that ACC1 mRNA is overexpressed in non-small-cell lung cancer (NSCLC), which is accompanied by reduced DNA methylation at CpG island S shore of ACC1. Our study further demonstrated that higher ACC1 levels are associated with poor prognosis in NSCLC patients. Besides, we developed a novel synthetic route for preparation of a known ACC inhibitor ND-646, synthesized a series of its derivatives and evaluated their activity against the enzyme ACC1 and the A549 cell. As results, most of the tested compounds showed potent ACC1 inhibitory activity with IC50 values 3-10 nM. Among them, compounds A2, A7 and A9 displayed strong cancer inhibitory activity with IC50 values 9-17 nM by impairing cell growth and inducing cell death. Preliminary SAR analysis clearly suggested that (R)-configuration and amide group were vital to ACC1 and A549 inhibition, since compound (S)-A1 (the enantiomer of ND-646) had poor activity of ACC1 inhibition and the carboxylic acid ND-630 almost lost anticancer effect on A549 cells. Collectively, these findings indicate that ACC1 is a potential biomarker and target for non-small-cell lung cancer, and ND-646 and its derivatives as ACC1 inhibitors deserve further study for treatment of NSCLC.
Collapse
Affiliation(s)
- En-Qin Li
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease and State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjia Xiang, Nanjing 210009, China
| | - Wei Zhao
- Department of Clinical Biochemistry, School of Laboratory Medicine, Chengdu Medical College, Chengdu 610050, China; Department of Respiratory Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610050, China
| | - Chenxi Zhang
- Central Laboratory, Nanjing Chest Hospital, Medical School of Southeast University, Nanjing, Jiangsu Province 210029, China
| | - Lu-Zhe Qin
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease and State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjia Xiang, Nanjing 210009, China
| | - Sheng-Jie Liu
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease and State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjia Xiang, Nanjing 210009, China
| | - Zhi-Qi Feng
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease and State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjia Xiang, Nanjing 210009, China
| | - Xiaoan Wen
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease and State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjia Xiang, Nanjing 210009, China.
| | - Cai-Ping Chen
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease and State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjia Xiang, Nanjing 210009, China.
| |
Collapse
|
23
|
Chen CP, Sang Y, Liu L, Feng ZQ, Liang Z, Pei X. THAP7 promotes cell proliferation by regulating the G1/S phase transition via epigenetically silencing p21 in lung adenocarcinoma. Onco Targets Ther 2019; 12:5651-5660. [PMID: 31372002 PMCID: PMC6634299 DOI: 10.2147/ott.s208908] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 07/03/2019] [Indexed: 12/21/2022] Open
Abstract
PURPOSE Lung adenocarcinoma (LUAD) is one of the most common cancers worldwide. The THanatos-Associated Proteins (THAP) family plays an essential role in multiple cancers. However, the role of THAP7 in cancers has remained elusive. METHODS THAP7 expression status in LUAD tissues was analysed by using the Oncomine database and qRT-PCR, and its expression level in LUAD cell lines was detected by qRT-PCR and Western blotting. The role of THAP7 in LUAD cells was determined by proliferation, colony formation, and cell cycle analyses. In vivo role of THAP7 was studied on xenograft models. Luciferase reporter assays and chromatin immunoprecipitation (ChIP) were used to determine the activity and acetylation of the p21 promoter. RESULTS THAP7 expression was increased in LUAD tissues and cell lines. Moreover, the high expression of THAP7 was correlated with poor prognosis. The overexpression of THAP7 accelerated the G1/S phase transition and promoted tumour growth both in vitro and in vivo. A mechanistic study revealed that THAP7 reduced the acetylation of histone H3 on the p21 promoter to suppress p21 transcription. CONCLUSION For the first time, we demonstrated the function of THAP7 in LUAD, and our findings suggested that THAP7 may be a potential molecular therapy target in LUAD.
Collapse
Affiliation(s)
- Cai-Ping Chen
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing210009, People’s Republic of China
| | - Yi Sang
- Department of Center Laboratory, Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, The Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi330008, People’s Republic of China
| | - Lijuan Liu
- Department of Pharmacy, Jiangxi Cancer Hospital, Nanchang, Jiangxi330029, People’s Republic of China
| | - Zhi-Qi Feng
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing210009, People’s Republic of China
| | - Zibin Liang
- Department of Thoracic Oncology, The Cancer Center of the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong519000, People’s Republic of China
| | - Xiaofeng Pei
- Department of Thoracic Oncology, The Cancer Center of the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong519000, People’s Republic of China
| |
Collapse
|
24
|
Xie Y, Perrino BA. Quantitative in situ proximity ligation assays examining protein interactions and phosphorylation during smooth muscle contractions. Anal Biochem 2019; 577:1-13. [PMID: 30981700 DOI: 10.1016/j.ab.2019.04.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/21/2019] [Accepted: 04/10/2019] [Indexed: 12/26/2022]
Abstract
Antibody-based in situ proximity ligation assays (isPLA) have the potential to study protein phosphorylation and protein interactions with spatial resolution in intact tissues. However, the application of isPLA at the tissue level is limited by a lack of appropriate positive and negative controls and the difficulty in accounting for changes in tissue shape. Here we demonstrate a set of experimental and computational approaches using gastric fundus smooth muscles to improve the validity of quantitative isPLA. Appropriate positive and negative biological controls and PLA technical controls were selected to ensure experimental rigor. To account for changes in morphology between relaxed and contracted smooth muscles, target PLA spots were normalized to smooth muscle myosin light chain 20 PLA spots or the cellular cross-sectional areas. We describe the computational steps necessary to filter out false-positive improperly sized spots and set the thresholds for counting true positive PLA spots to quantify the PLA signals. We tested our approach by examining protein phosphorylation and protein interactions in smooth muscle myofilament Ca2+ sensitization pathways from resting and contracted gastric fundus smooth muscles. In conclusion, our tissue-level isPLA method enables unbiased quantitation of protein phosphorylation and protein-protein interactions in intact smooth muscle tissues, suggesting the potential for quantitative isPLA applications in other types of intact tissues.
Collapse
Affiliation(s)
- Yeming Xie
- Department of Physiology and Cell Biology, University of Nevada Reno, School of Medicine, MS 0352, 1664 N Virginia St, Reno, NV, 89557, USA
| | - Brian A Perrino
- Department of Physiology and Cell Biology, University of Nevada Reno, School of Medicine, MS 0352, 1664 N Virginia St, Reno, NV, 89557, USA.
| |
Collapse
|
25
|
Mason DE, Collins JM, Dawahare JH, Nguyen TD, Lin Y, Voytik-Harbin SL, Zorlutuna P, Yoder MC, Boerckel JD. YAP and TAZ limit cytoskeletal and focal adhesion maturation to enable persistent cell motility. J Cell Biol 2019; 218:1369-1389. [PMID: 30737263 PMCID: PMC6446844 DOI: 10.1083/jcb.201806065] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 11/29/2018] [Accepted: 01/11/2019] [Indexed: 12/18/2022] Open
Abstract
Cell migration initiates by traction generation through reciprocal actomyosin tension and focal adhesion reinforcement, but continued motility requires adaptive cytoskeletal remodeling and adhesion release. Here, we asked whether de novo gene expression contributes to this cytoskeletal feedback. We found that global inhibition of transcription or translation does not impair initial cell polarization or migration initiation, but causes eventual migratory arrest through excessive cytoskeletal tension and over-maturation of focal adhesions, tethering cells to their matrix. The transcriptional coactivators YAP and TAZ mediate this feedback response, modulating cell mechanics by limiting cytoskeletal and focal adhesion maturation to enable persistent cell motility and 3D vasculogenesis. Motile arrest after YAP/TAZ ablation was partially rescued by depletion of the YAP/TAZ-dependent myosin phosphatase regulator, NUAK2, or by inhibition of Rho-ROCK-myosin II. Together, these data establish a transcriptional feedback axis necessary to maintain a responsive cytoskeletal equilibrium and persistent migration.
Collapse
Affiliation(s)
- Devon E Mason
- McKay Orthopaedic Research Laboratory, Department of Orthopedic Surgery, University of Pennsylvania, Philadelphia, PA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN
| | - Joseph M Collins
- McKay Orthopaedic Research Laboratory, Department of Orthopedic Surgery, University of Pennsylvania, Philadelphia, PA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA
| | - James H Dawahare
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN
| | - Trung Dung Nguyen
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN
- Department of Engineering and Computer Science, Seattle Pacific University, Seattle, WA
| | - Yang Lin
- Herman B. Wells Center for Pediatric Research, Indiana University, Indianapolis, IN
| | - Sherry L Voytik-Harbin
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN
| | - Pinar Zorlutuna
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN
| | - Mervin C Yoder
- Herman B. Wells Center for Pediatric Research, Indiana University, Indianapolis, IN
| | - Joel D Boerckel
- McKay Orthopaedic Research Laboratory, Department of Orthopedic Surgery, University of Pennsylvania, Philadelphia, PA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN
| |
Collapse
|
26
|
CPI-17-mediated contraction of vascular smooth muscle is essential for the development of hypertension in obese mice. J Genet Genomics 2019; 46:109-118. [PMID: 30948334 DOI: 10.1016/j.jgg.2019.02.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 01/21/2019] [Accepted: 02/20/2019] [Indexed: 12/14/2022]
Abstract
Several factors have been implicated in obesity-related hypertension, but the genesis of the hypertension is largely unknown. In this study, we found a significantly upregulated expression of CPI-17 (C-kinase-potentiated protein phosphatase 1 inhibitor of 17 kDa) and protein kinase C (PKC) isoforms in the vascular smooth muscles of high-fat diet (HFD)-fed obese mice. The obese wild-type mice showed a significant elevation of blood pressure and enhanced calcium-sensitized contraction of vascular smooth muscles. However, the obese CPI-17-deficient mice showed a normotensive blood pressure, and the calcium-sensitized contraction was consistently reduced. In addition, the mutant muscle displayed an abolished responsive force to a PKC activator and a 30%-50% reduction in both the initial peak force and sustained force in response to various G protein-coupled receptor (GPCR) agonists. Our observations showed that CPI-17-mediated calcium sensitization is mediated through a GPCR/PKC/CPI-17/MLCP/RLC signaling pathway. We therefore propose that the upregulation of CPI-17-mediated calcium-sensitized vasocontraction by obesity contributes to the development of obesity-related hypertension.
Collapse
|
27
|
Lubomirov LT, Gagov H, Schroeter MM, Wiesner RJ, Franko A. Augmented contractility of murine femoral arteries in a streptozotocin diabetes model is related to increased phosphorylation of MYPT1. Physiol Rep 2019; 7:e13975. [PMID: 30740930 PMCID: PMC6369311 DOI: 10.14814/phy2.13975] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 12/13/2018] [Indexed: 12/19/2022] Open
Abstract
Diabetes mellitus (DM) is a metabolic disorder with high prevalence, and a major risk factor for macro- and microvascular abnormalities. This study was undertaken to explore the mechanisms of hypercontractility of murine femoral arteries (FA) obtained from mice with streptozotocin (STZ)-induced diabetes and its relation to the phosphorylation profile of the myosin phosphatase target subunit 1, MYPT1. The immunoreactivity of MYPT1 toward phospho-MYPT1-T696, MYPT1-T853, or MYPT1-S695, used as a read out for MYPT1 phosphorylation, has been studied by Western Blotting. Contractile activity of FA from control and STZ mice has been studied by wire myography. At basal conditions (no treatment), the immunoreactivity of MYPT1-T696/T853 was ~2-fold higher in the STZ arteries compared with controls. No changes in MYPT1-T696/853 phosphorylation were observed after stimulation with the Thromboxan-A2 analog, U46619. Neither basal nor U46619-stimulated phosphorylation of MYPT1 at S695 was affected by STZ treatment. Mechanical distensibility and basal tone of FA obtained from STZ animals were similar to controls. Maximal force after treatment of FA with the contractile agonists phenylephrine (10 μmol/L) or U46619 (1 μmol/L) was augmented in the arteries of STZ mice by ~2- and ~1.5-fold, respectively. In summary, our study suggests that development of a hypercontractile phenotype in murine FA in STZ diabetes is at least partially related to an increase in phosphorylation of MLCP at MYPT1-T696/853. Interestingly, the phosphorylation at S695 site was not altered in STZ-induced diabetes, supporting the view that S695 may serve as a sensor for mechanical activity which is not directly involved in tone regulation.
Collapse
Affiliation(s)
| | - Hristo Gagov
- Faculty of BiologySofia University St. Kliment OhridskiSofiaBulgaria
| | | | - Rudolf J. Wiesner
- Institute of Vegetative PhysiologyUniversity of CologneKölnGermany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD)KölnGermany
| | - Andras Franko
- Institute of Vegetative PhysiologyUniversity of CologneKölnGermany
- German Center for Diabetes Research (DZD e.V.)NeuherbergGermany
- Division of EndocrinologyDepartment of Internal Medicine IVDiabetology, Angiology, Nephrology and Clinical ChemistryUniversity of TübingenTübingenGermany
| |
Collapse
|
28
|
George Z, Omosun Y, Azenabor AA, Goldstein J, Partin J, Joseph K, Ellerson D, He Q, Eko F, McDonald MA, Reed M, Svoboda P, Stuchlik O, Pohl J, Lutter E, Bandea C, Black CM, Igietseme JU. The molecular mechanism of induction of unfolded protein response by Chlamydia. Biochem Biophys Res Commun 2019; 508:421-429. [PMID: 30503337 PMCID: PMC6343654 DOI: 10.1016/j.bbrc.2018.11.034] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 11/06/2018] [Indexed: 11/21/2022]
Abstract
The unfolded protein response (UPR) contributes to chlamydial pathogenesis, as a source of lipids and ATP during replication, and for establishing the initial anti-apoptotic state of host cell that ensures successful inclusion development. The molecular mechanism(s) of UPR induction by Chlamydia is unknown. Chlamydia use type III secretion system (T3SS) effector proteins (e.g, the Translocated Actin-Recruiting Phosphoprotein (Tarp) to stimulate host cell's cytoskeletal reorganization that facilitates invasion and inclusion development. We investigated the hypothesis that T3SS effector-mediated assembly of myosin-II complex produces activated non-muscle myosin heavy chain II (NMMHC-II), which then binds the UPR master regulator (BiP) and/or transducers to induce UPR. Our results revealed the interaction of the chlamydial effector proteins (CT228 and Tarp) with components of the myosin II complex and UPR regulator and transducer during infection. These interactions caused the activation and binding of NMMHC-II to BiP and IRE1α leading to UPR induction. In addition, specific inhibitors of myosin light chain kinase, Tarp oligomerization and myosin ATPase significantly reduced UPR activation and Chlamydia replication. Thus, Chlamydia induce UPR through T3SS effector-mediated activation of NMMHC-II components of the myosin complex to facilitate infectivity. The finding provides greater insights into chlamydial pathogenesis with the potential to identify therapeutic targets and formulations.
Collapse
Affiliation(s)
- Zenas George
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | - Yusuf Omosun
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA; Morehouse School of Medicine, Atlanta, GA, USA
| | | | - Jason Goldstein
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | - James Partin
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | - Kahaliah Joseph
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | - Debra Ellerson
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | - Qing He
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA; Morehouse School of Medicine, Atlanta, GA, USA
| | - Francis Eko
- Morehouse School of Medicine, Atlanta, GA, USA
| | | | - Matthew Reed
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | - Pavel Svoboda
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | - Olga Stuchlik
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | - Jan Pohl
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | | | - Claudiu Bandea
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | - Carolyn M Black
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | - Joseph U Igietseme
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA; Morehouse School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
29
|
Chang AN, Gao N, Liu Z, Huang J, Nairn AC, Kamm KE, Stull JT. The dominant protein phosphatase PP1c isoform in smooth muscle cells, PP1cβ, is essential for smooth muscle contraction. J Biol Chem 2018; 293:16677-16686. [PMID: 30185619 PMCID: PMC6204911 DOI: 10.1074/jbc.ra118.003083] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 08/30/2018] [Indexed: 12/29/2022] Open
Abstract
Contractile force development of smooth muscle is controlled by balanced kinase and phosphatase activities toward the myosin regulatory light chain (RLC). Numerous biochemical and pharmacological studies have investigated the specificity and regulatory activity of smooth muscle myosin light-chain phosphatase (MLCP) bound to myosin filaments and comprised of the regulatory myosin phosphatase target subunit 1 (MYPT1) and catalytic protein phosphatase 1cβ (PP1cβ) subunits. Recent physiological and biochemical evidence obtained with smooth muscle tissues from a conditional MYPT1 knockout suggests that a soluble, MYPT1-unbound form of PP1cβ may additionally contribute to myosin RLC dephosphorylation and relaxation of smooth muscle. Using a combination of isoelectric focusing and isoform-specific immunoblotting, we found here that more than 90% of the total PP1c in mouse smooth muscles is the β isoform. Moreover, conditional knockout of PP1cα or PP1cγ in adult smooth muscles did not result in an apparent phenotype in mice up to 6 months of age and did not affect smooth muscle contractions ex vivo In contrast, smooth muscle-specific conditional PP1cβ knockout decreased contractile force development in bladder, ileal, and aortic tissues and reduced mouse survival. Bladder smooth muscle tissue from WT mice was selectively permeabilized to remove soluble PP1cβ to measure contributions of total (α-toxin treatment) and myosin-bound (Triton X-100 treatment) phosphatase activities toward phosphorylated RLC in myofilaments. Triton X-100 reduced PP1cβ content by 60% and the rate of RLC dephosphorylation by 2-fold. These results are consistent with the selective dephosphorylation of RLC by both MYPT1-bound and -unbound PP1cβ forms in smooth muscle.
Collapse
Affiliation(s)
- Audrey N Chang
- From the Departments of Physiology and
- Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9040 and
| | - Ning Gao
- From the Departments of Physiology and
| | | | | | - Angus C Nairn
- the Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut 06508
| | | | | |
Collapse
|
30
|
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
|
31
|
Mochalov SV, Tarasova NV, Kudryashova TV, Gaynullina DK, Kalenchuk VU, Borovik AS, Vorotnikov AV, Tarasova OS, Schubert R. Higher Ca 2+ -sensitivity of arterial contraction in 1-week-old rats is due to a greater Rho-kinase activity. Acta Physiol (Oxf) 2018; 223:e13044. [PMID: 29383848 DOI: 10.1111/apha.13044] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 01/23/2018] [Accepted: 01/24/2018] [Indexed: 12/22/2022]
Abstract
AIM During early post-natal development, arterial contraction depends less on Ca2+ -signalling pathways but more on changes in Ca2+ -sensitivity compared to adult animals. Whether this difference is related to Rho-kinase, one of the major players affecting Ca2+ -sensitivity, is unknown for intact vessels. Thus, we tested the hypothesis that Rho-kinase critically contributes to the higher Ca2+ -sensitivity of contraction in intact arteries of 1-week-old rats. METHODS We studied 1-week-old, 4- to 5-week-old and 10- to 12-week-old rats performing isometric myography, Ca2+ -fluorimetry and Western blotting using intact saphenous arteries and arterial pressure measurements under urethane anaesthesia. RESULTS In 10- to 12-week-old rats, methoxamine (MX) produced vasoconstriction associated with an increase in [Ca2+ ]i and Ca2+ -sensitivity. In contrast, in 1-week-old rats these contractions were accompanied only by an increase in Ca2+ -sensitivity. All MX-induced effects were reduced by the Rho-kinase inhibitor Y-27632; this reduction was complete only in 1-week-old rats. The Rho-kinase specific site Thr855 on MYPT1 was increasingly phosphorylated by MX in vessels of 1-week-old, but not 10- to 12-week-old rats; this effect was also inhibited completely by Y-27632. The Rho-kinase inhibitor fasudil in a dose not affecting the pressor response to MX in 4- to 5-week-old rats reduced it considerably in 1-week-old rats. CONCLUSION Our results suggest that the higher Ca2+ -sensitivity of arterial contraction in 1-week-old compared to 10- to 12-week-old rats is due to a greater Rho-kinase activity. Constitutively active Rho-kinase contributes to MX-induced contraction in 10- to 12-week-old rats. In 1-week-old rats, additional Rho-kinase activation is involved. This remodelling of the Rho-kinase pathway is associated with its increased contribution to adrenergic arterial pressure responses.
Collapse
Affiliation(s)
- S. V. Mochalov
- Medical Faculty Mannheim; Centre for Biomedicine and Medical Technology Mannheim (CBTM) and European Center of Angioscience (ECAS); Research Division Cardiovascular Physiology; Heidelberg University; Mannheim Germany
- Faculty of Biology, M.V. Lomonosov; Moscow State University; Moscow Russia
- ChemRar Research and Development Institute; Khimki Moscow Region Russia
| | - N. V. Tarasova
- Medical Faculty Mannheim; Centre for Biomedicine and Medical Technology Mannheim (CBTM) and European Center of Angioscience (ECAS); Research Division Cardiovascular Physiology; Heidelberg University; Mannheim Germany
- Faculty of Biology, M.V. Lomonosov; Moscow State University; Moscow Russia
- Molecular Medicine Institute; I.M. Sechenov First Moscow State Medical University; Moscow Russia
| | - T. V. Kudryashova
- Institute of Experimental Cardiology; Cardiology Research Center; Moscow Russia
| | - D. K. Gaynullina
- Medical Faculty Mannheim; Centre for Biomedicine and Medical Technology Mannheim (CBTM) and European Center of Angioscience (ECAS); Research Division Cardiovascular Physiology; Heidelberg University; Mannheim Germany
- Faculty of Biology, M.V. Lomonosov; Moscow State University; Moscow Russia
- Department of Physiology; Russian National Research Medical University; Moscow Russia
| | - V. U. Kalenchuk
- Faculty of Basic Medicine; M.V. Lomonosov Moscow State University; Moscow Russia
| | - A. S. Borovik
- State Research Center of the Russian Federation - Institute for Biomedical Problems; Russian Academy of Sciences; Moscow Russia
| | - A. V. Vorotnikov
- Institute of Experimental Cardiology; Cardiology Research Center; Moscow Russia
- Medical Center; M.V. Lomonosov Moscow State University; Moscow Russia
| | - O. S. Tarasova
- Faculty of Biology, M.V. Lomonosov; Moscow State University; Moscow Russia
- State Research Center of the Russian Federation - Institute for Biomedical Problems; Russian Academy of Sciences; Moscow Russia
| | - R. Schubert
- Medical Faculty Mannheim; Centre for Biomedicine and Medical Technology Mannheim (CBTM) and European Center of Angioscience (ECAS); Research Division Cardiovascular Physiology; Heidelberg University; Mannheim Germany
| |
Collapse
|
32
|
Xie Y, Han KH, Grainger N, Li W, Corrigan RD, Perrino BA. A role for focal adhesion kinase in facilitating the contractile responses of murine gastric fundus smooth muscles. J Physiol 2018. [PMID: 29528115 DOI: 10.1113/jp275406] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
KEY POINTS Activation of focal adhesion kinase (FAK) by integrin signalling facilitates smooth muscle contraction by transmitting the force generated by myofilament activation to the extracellular matrix and throughout the smooth muscle tissue. Here we report that electrical field stimulation (EFS) of cholinergic motor neurons activates FAK in gastric fundus smooth muscles, and that FAK activation by EFS is atropine-sensitive but nicardipine-insensitive. PDBu and calyculin A contracted gastric fundus muscles Ca2+ -independently and also activated FAK. Inhibition of FAK activation inhibits the contractile responses evoked by EFS, and inhibits CPI-17 phosphorylation at T38. This study indicates that mechanical force or tension is sufficient to activate FAK, and that FAK appears to be involved in the activation of the protein kinase C-CPI-17 Ca2+ sensitization pathway in gastric fundus smooth muscles. These results reveal a novel role for FAK in gastric fundus smooth muscle contraction by facilitating CPI-17 phosphorylation. ABSTRACT Smooth muscle contraction involves regulating myosin light chain phosphorylation and dephosphorylation by myosin light chain kinase and myosin light chain phosphatase. C-kinase potentiated protein phosphatase-1 inhibitor of 17 kDa (CPI-17) and myosin phosphatase targeting subunit of myosin light-chain phosphatase (MYPT1) are crucial for regulating gastrointestinal smooth muscle contraction by inhibiting myosin light chain phosphatase. Integrin signalling involves the dynamic recruitment of several proteins, including focal adhesion kinase (FAK), to focal adhesions. FAK tyrosine kinase activation is involved in cell adhesion to the extracellular matrix via integrin signalling. FAK participates in linking the force generated by myofilament activation to the extracellular matrix and throughout the smooth muscle tissue. Here, we show that cholinergic stimulation activates FAK in gastric fundus smooth muscles. Electrical field stimulation in the presence of Nω -nitro-l-arginine methyl ester and MRS2500 contracted gastric fundus smooth muscle strips and increased FAK Y397 phosphorylation (pY397). Atropine blocked the contractions and prevented the increase in pY397. The FAK inhibitor PF-431396 inhibited the contractions and the increase in pY397. PF-431396 also inhibited the electrical field stimulation-induced increase in CPI-17 T38 phosphorylation, and reduced MYPT1 T696 and T853, and myosin light chain S19 phosphorylation. Ca2+ influx was unaffected by PF-431396. Nicardipine inhibited the contractions but had no effect on the increase in pY397. Phorbol 12,13-dibutyrate or calyculin A contracted gastric fundus smooth muscle strips Ca2+ independently and increased pY397. Our findings suggest that FAK is activated by mechanical forces during contraction and reveal a novel role of FAK in the regulation of CPI-17 phosphorylation.
Collapse
Affiliation(s)
- Yeming Xie
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, NV, 89557, USA
| | - Koon Hee Han
- Department of Internal Medicine, Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung, Republic of Korea
| | - Nathan Grainger
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, NV, 89557, USA
| | - Wen Li
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, NV, 89557, USA
| | - Robert D Corrigan
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, NV, 89557, USA
| | - Brian A Perrino
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, NV, 89557, USA
| |
Collapse
|
33
|
Yang Q, Fujii W, Kaji N, Kakuta S, Kada K, Kuwahara M, Tsubone H, Ozaki H, Hori M. The essential role of phospho‐T38 CPI‐17 in the maintenance of physiological blood pressure using genetically modified mice. FASEB J 2018; 32:2095-2109. [DOI: 10.1096/fj.201700794r] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Qunhui Yang
- Department of Veterinary Pharmacology, Laboratory of Applied Genetics, Department of Biomedical Science, Department of Veterinary Pathophysiology and Animal Health, and Research Center for Food SafetyGraduate School of Agriculture and Life Sciences, The University of TokyoTokyoJapan
| | - Wataru Fujii
- Laboratory of Applied Genetics, Department of Biomedical Science, Department of Veterinary Pathophysiology and Animal Health, and Research Center for Food SafetyGraduate School of Agriculture and Life Sciences, The University of TokyoTokyoJapan
| | - Noriyuki Kaji
- Department of Veterinary Pharmacology, Laboratory of Applied Genetics, Department of Biomedical Science, Department of Veterinary Pathophysiology and Animal Health, and Research Center for Food SafetyGraduate School of Agriculture and Life Sciences, The University of TokyoTokyoJapan
| | - Shigeru Kakuta
- Department of Biomedical Science, Department of Veterinary Pathophysiology and Animal Health, and Research Center for Food SafetyGraduate School of Agriculture and Life Sciences, The University of TokyoTokyoJapan
| | - Kodai Kada
- Department of Veterinary Pharmacology, Laboratory of Applied Genetics, Department of Biomedical Science, Department of Veterinary Pathophysiology and Animal Health, and Research Center for Food SafetyGraduate School of Agriculture and Life Sciences, The University of TokyoTokyoJapan
| | - Masayoshi Kuwahara
- Department of Veterinary Pathophysiology and Animal Health, and Research Center for Food SafetyGraduate School of Agriculture and Life Sciences, The University of TokyoTokyoJapan
| | - Hirokazu Tsubone
- Research Center for Food SafetyGraduate School of Agriculture and Life Sciences, The University of TokyoTokyoJapan
| | - Hiroshi Ozaki
- Department of Veterinary Pharmacology, Laboratory of Applied Genetics, Department of Biomedical Science, Department of Veterinary Pathophysiology and Animal Health, and Research Center for Food SafetyGraduate School of Agriculture and Life Sciences, The University of TokyoTokyoJapan
| | - Masatoshi Hori
- Department of Veterinary Pharmacology, Laboratory of Applied Genetics, Department of Biomedical Science, Department of Veterinary Pathophysiology and Animal Health, and Research Center for Food SafetyGraduate School of Agriculture and Life Sciences, The University of TokyoTokyoJapan
| |
Collapse
|
34
|
Eto M, Kitazawa T. Diversity and plasticity in signaling pathways that regulate smooth muscle responsiveness: Paradigms and paradoxes for the myosin phosphatase, the master regulator of smooth muscle contraction. J Smooth Muscle Res 2018; 53:1-19. [PMID: 28260704 PMCID: PMC5364378 DOI: 10.1540/jsmr.53.1] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
A hallmark of smooth muscle cells is their ability to adapt their functions to meet temporal and chronic fluctuations in their demands. These functions include force development and growth. Understanding the mechanisms underlying the functional plasticity of smooth muscles, the major constituent of organ walls, is fundamental to elucidating pathophysiological rationales of failures of organ functions. Also, the knowledge is expected to facilitate devising innovative strategies that more precisely monitor and normalize organ functions by targeting individual smooth muscles. Evidence has established a current paradigm that the myosin light chain phosphatase (MLCP) is a master regulator of smooth muscle responsiveness to stimuli. Cellular MLCP activity is negatively and positively regulated in response to G-protein activation and cAMP/cGMP production, respectively, through the MYPT1 regulatory subunit and an endogenous inhibitor protein named CPI-17. In this article we review the outcomes from two decade of research on the CPI-17 signaling and discuss emerging paradoxes in the view of signaling pathways regulating smooth muscle functions through MLCP.
Collapse
Affiliation(s)
- Masumi Eto
- Department of Molecular Physiology and Biophysics, Sidney Kimmel Medical College at Thomas Jefferson University and Sidney Kimmel Cancer Center, 1020 Locust Street, Philadelphia, PA19107, USA
| | | |
Collapse
|
35
|
Li W, Sasse KC, Bayguinov Y, Ward SM, Perrino BA. Contractile Protein Expression and Phosphorylation and Contractility of Gastric Smooth Muscles from Obese Patients and Patients with Obesity and Diabetes. J Diabetes Res 2018; 2018:8743874. [PMID: 29955616 PMCID: PMC6000859 DOI: 10.1155/2018/8743874] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 04/11/2018] [Accepted: 04/18/2018] [Indexed: 12/22/2022] Open
Abstract
Ingested food is received, mixed, and ground into chyme by distinct gastric motility patterns. Diabetes impairs gastric muscle function, but the mechanisms underlying diabetes-induced gastric muscle dysfunction are unknown. Here, we compared the expression and phosphorylation of Ca2+ sensitization and contractile proteins in human gastric muscles from obese nondiabetic and diabetic patients. We also compared the spontaneous phasic contractions and the contractile responses evoked by electrical field stimulation of cholinergic motor neurons. Fundus and antrum muscles were obtained from sleeve gastrectomies and were used in in vitro myobath contractile studies and for capillary electrophoresis and immunodetection of γ-actin, CPI-17, pT38-CPI-17, MYPT1, pT853-MYPT1, pT696-MYPT1, myosin light chain (MYL9), pS19-MYL9, myosin light chain kinase (MYLK), protein phosphatase-1δ (PP1δ), and Rho-associated kinase (ROCK2). In diabetic fundus muscles, MYLK, ROCK2, and PP1δ expression was unchanged; MYPT1 and CPI-17 expression was decreased; and the pT853/MYPT1 and pT38/CPI-17 ratios, but not the pT696/MYPT1 ratio, were increased. Although MYL9 expression was increased, the pS19/MYL9 ratio was unchanged in diabetic fundus muscles. In diabetic antrum muscles, MYLK and MYL9 expression was unchanged, but ROCK2, CPI-17, and PP1δ expression was decreased. The pT38/CPI-17 ratio was unchanged, while the pS19/MYL9, pT853/MYPT1, and pT696/MYPT1 ratios were decreased, consistent with the reduced ROCK2 expression. The frequencies of spontaneous phasic contractions from nondiabetic and diabetic gastric fundus and antrum muscles did not significantly differ from each other, regardless of age, sex, or diabetic status. The fold increases in the contractions of diabetic fundus and antrum muscles in response to increased frequencies of electrical field stimulation were significantly lower compared to nondiabetic fundus and antrum muscles. The altered contractile responses and the protein expression and phosphorylation in gastric muscles of obese patients with diabetes illustrate the importance of understanding how smooth muscle Ca2+ sensitization mechanisms contribute to gastric motility.
Collapse
Affiliation(s)
- Wen Li
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
| | - Kent C. Sasse
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
- Sasse Surgical Associates, Reno NV 89502, USA
- Renown Regional Medical Center, Reno, NV 89502, USA
| | - Yulia Bayguinov
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
| | - Sean M. Ward
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
| | - Brian A. Perrino
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
| |
Collapse
|
36
|
Takai A, Eto M, Hirano K, Takeya K, Wakimoto T, Watanabe M. Protein phosphatases 1 and 2A and their naturally occurring inhibitors: current topics in smooth muscle physiology and chemical biology. J Physiol Sci 2018; 68:1-17. [PMID: 28681362 PMCID: PMC5754374 DOI: 10.1007/s12576-017-0556-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 06/27/2017] [Indexed: 12/26/2022]
Abstract
Protein phosphatases 1 and 2A (PP1 and PP2A) are the most ubiquitous and abundant serine/threonine phosphatases in eukaryotic cells. They play fundamental roles in the regulation of various cellular functions. This review focuses on recent advances in the functional studies of these enzymes in the field of smooth muscle physiology. Many naturally occurring protein phosphatase inhibitors with different relative PP1/PP2A affinities have been discovered and are widely used as powerful research tools. Current topics in the chemical biology of PP1/PP2A inhibitors are introduced and discussed, highlighting the identification of the gene cluster responsible for the biosynthesis of calyculin A in a symbiont microorganism of a marine sponge.
Collapse
Affiliation(s)
- Akira Takai
- Department of Physiology, Asahikawa Medical University, Midorigaoka-Higashi 2-1-1-1, Asahikwa, 078-8510, Japan.
| | - Masumi Eto
- Department of Molecular Physiology and Biophysics and Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Katsuya Hirano
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| | - Kosuke Takeya
- Department of Physiology, Asahikawa Medical University, Midorigaoka-Higashi 2-1-1-1, Asahikwa, 078-8510, Japan
| | - Toshiyuki Wakimoto
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12, Nishi 6, Kita-ku, Sapporo, 060-0812, Japan
| | - Masaru Watanabe
- Department of Frontier Health Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Higashi-Ogu, Arakawa-ku 7-2-10, Tokyo, 116-8551, Japan
| |
Collapse
|
37
|
Gao N, Tsai MH, Chang AN, He W, Chen CP, Zhu M, Kamm KE, Stull JT. Physiological vs. pharmacological signalling to myosin phosphorylation in airway smooth muscle. J Physiol 2017; 595:6231-6247. [PMID: 28749013 PMCID: PMC5621497 DOI: 10.1113/jp274715] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 07/25/2017] [Indexed: 01/05/2023] Open
Abstract
KEY POINTS Smooth muscle myosin regulatory light chain (RLC) is phosphorylated by Ca2+ /calmodulin-dependent myosin light chain kinase and dephosphorylated by myosin light chain phosphatase (MLCP). Tracheal smooth muscle contains significant amounts of myosin binding subunit 85 (MBS85), another myosin phosphatase targeting subunit (MYPT) family member, in addition to MLCP regulatory subunit MYPT1. Concentration/temporal responses to carbachol demonstrated similar sensitivities for bovine tracheal force development and phosphorylation of RLC, MYPT1, MBS85 and paxillin. Electrical field stimulation releases ACh from nerves to increase RLC phosphorylation but not MYPT1 or MBS85 phosphorylation. Thus, nerve-mediated muscarinic responses in signalling modules acting on RLC phosphorylation are different from pharmacological responses with bath added agonist. The conditional knockout of MYPT1 or the knock-in mutation T853A in mice had no effect on muscarinic force responses in isolated tracheal tissues. MLCP activity may arise from functionally shared roles between MYPT1 and MBS85, resulting in minimal effects of MYPT1 knockout on contraction. ABSTRACT Ca2+ /calmodulin activation of myosin light chain kinase (MLCK) initiates myosin regulatory light chain (RLC) phosphorylation for smooth muscle contraction with subsequent dephosphorylation for relaxation by myosin light chain phosphatase (MLCP) containing regulatory (MYPT1) and catalytic (PP1cδ) subunits. RLC phosphorylation-dependent force development is regulated by distinct signalling modules involving protein phosphorylations. We investigated responses to cholinergic agonist treatment vs. neurostimulation by electric field stimulation (EFS) in bovine tracheal smooth muscle. Concentration/temporal responses to carbachol demonstrated tight coupling between force development and RLC phosphorylation but sensitivity differences in MLCK, MYPT1 T853, MYPT1 T696, myosin binding subunit 85 (MBS85), paxillin and CPI-17 (PKC-potentiated protein phosphatase 1 inhibitor protein of 17 kDa) phosphorylations. EFS increased force and phosphorylation of RLC, CPI-17 and MLCK. In the presence of the cholinesterase inhibitor neostigmine, EFS led to an additional increase in phosphorylation of MYPT1 T853, MYPT1 T696, MBS85 and paxillin. Thus, there were distinct pharmacological vs. physiological responses in signalling modules acting on RLC phosphorylation and force responses, probably related to degenerate G protein signalling networks. Studies with genetically modified mice were performed. Expression of another MYPT1 family member, MBS85, was enriched in mouse, as well as bovine tracheal smooth muscle. Carbachol concentration/temporal-force responses were similar in trachea from MYPT1SM+/+ , MYPT1SM-/- and the knock-in mutant mice containing nonphosphorylatable MYPT1 T853A with no differences in RLC phosphorylation. Thus, MYPT1 T853 phosphorylation was not necessary for regulation of RLC phosphorylation in tonic airway smooth muscle. Furthermore, MLCP activity may arise from functionally shared roles between MYPT1 and MBS85, resulting in minimal effects of MYPT1 knockout on contraction.
Collapse
Affiliation(s)
- Ning Gao
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ming-Ho Tsai
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Present address: Graduate Institute of Medicine, Kaohsiung Medical University, 100 Shih-Chuan 1st Rd, San Ming District, Kaohsiung, Taiwan
| | - Audrey N Chang
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Weiqi He
- Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing, China.,Present address: Cambridge-Suda (CAM-SU) Genomic Resource Center, Soochow University, Suzhou, China
| | - Cai-Ping Chen
- Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing, China.,Present address: Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing, PR China
| | - Minsheng Zhu
- Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing, China
| | - Kristine E Kamm
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - James T Stull
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
38
|
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
|
39
|
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
|
40
|
Lubomirov LT, Papadopoulos S, Pütz S, Welter J, Klöckener T, Weckmüller K, Ardestani MA, Filipova D, Metzler D, Metzner H, Staszewski J, Zittrich S, Gagov H, Schroeter MM, Pfitzer G. Aging-related alterations in eNOS and nNOS responsiveness and smooth muscle reactivity of murine basilar arteries are modulated by apocynin and phosphorylation of myosin phosphatase targeting subunit-1. J Cereb Blood Flow Metab 2017; 37:1014-1029. [PMID: 27193035 PMCID: PMC5363478 DOI: 10.1177/0271678x16649402] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 04/07/2016] [Accepted: 04/08/2016] [Indexed: 12/20/2022]
Abstract
Aging causes major alterations of all components of the neurovascular unit and compromises brain blood supply. Here, we tested how aging affects vascular reactivity in basilar arteries from young (<10 weeks; y-BA), old (>22 months; o-BA) and old (>22 months) heterozygous MYPT1-T-696A/+ knock-in mice. In isometrically mounted o-BA, media thickness was increased by ∼10% while the passive length tension relations were not altered. Endothelial denudation or pan-NOS inhibition (100 µmol/L L-NAME) increased the basal tone by 11% in y-BA and 23% in o-BA, while inhibition of nNOS (1 µmol/L L-NPA) induced ∼10% increase in both ages. eNOS expression was ∼2-fold higher in o-BA. In o-BA, U46619-induced force was augmented (pEC50 ∼6.9 vs. pEC50 ∼6.5) while responsiveness to DEA-NONOate, electrical field stimulation or nicotine was decreased. Basal phosphorylation of MLC20-S19 and MYPT1-T-853 was higher in o-BA and was reversed by apocynin. Furthermore, permeabilized o-BA showed enhanced Ca2+-sensitivity. Old T-696A/+ BA displayed a reduced phosphorylation of MYPT1-T696 and MLC20, a lower basal tone in response to L-NAME and a reduced eNOS expression. The results indicate that the vascular hypercontractility found in o-BA is mediated by inhibition of MLCP and is partially compensated by an upregulation of endothelial NO release.
Collapse
Affiliation(s)
| | | | - Sandra Pütz
- Institute of Vegetative Physiology, University of Cologne, Germany
| | - Johannes Welter
- Institute of Vegetative Physiology, University of Cologne, Germany
| | - Tim Klöckener
- Institute for Genetics, University of Cologne, Germany
| | | | | | - Dilyana Filipova
- Institute of Vegetative Physiology, University of Cologne, Germany
| | - Doris Metzler
- Institute of Vegetative Physiology, University of Cologne, Germany
| | - Harald Metzner
- Institute of Vegetative Physiology, University of Cologne, Germany
| | | | - Stefan Zittrich
- Institute of Vegetative Physiology, University of Cologne, Germany
| | - Hristo Gagov
- Faculty of Biology, Sofia University St. Kliment Ohridski, Sofia, Bulgaria
| | | | - Gabriele Pfitzer
- Institute of Vegetative Physiology, University of Cologne, Germany
| |
Collapse
|
41
|
Smooth Muscle Phenotypic Diversity: Effect on Vascular Function and Drug Responses. ADVANCES IN PHARMACOLOGY 2017. [PMID: 28212802 DOI: 10.1016/bs.apha.2016.07.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
At its simplest resistance to blood flow is regulated by changes in the state of contraction of the vascular smooth muscle (VSM), a function of the competing activities of the myosin kinase and phosphatase determining the phosphorylation and activity of the myosin ATPase motor protein. In contrast, the vascular system of humans and other mammals is incredibly complex and highly regulated. Much of this complexity derives from phenotypic diversity within the smooth muscle, reflected in very differing power outputs and responses to signaling pathways that regulate vessel tone, presumably having evolved over the millennia to optimize vascular function and its control. The highly regulated nature of VSM tone, described as pharmacomechanical coupling, likely underlies the many classes of drugs in clinical use to alter vascular tone through activation or inhibition of these signaling pathways. This review will first describe the phenotypic diversity within VSM, followed by presentation of specific examples of how molecular diversity in signaling, myofilament, and calcium cycling proteins impacts arterial smooth muscle function and drug responses.
Collapse
|
42
|
Chang AN, Kamm KE, Stull JT. Role of myosin light chain phosphatase in cardiac physiology and pathophysiology. J Mol Cell Cardiol 2016; 101:35-43. [PMID: 27742556 DOI: 10.1016/j.yjmcc.2016.10.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 10/07/2016] [Accepted: 10/10/2016] [Indexed: 11/18/2022]
Abstract
Maintenance of contractile performance of the heart is achieved in part by the constitutive 40% phosphorylation of myosin regulatory light chain (RLC) in sarcomeres. The importance of this extent of RLC phosphorylation for optimal cardiac performance becomes apparent when various mouse models and resultant phenotypes are compared. The absence or attenuation of RLC phosphorylation results in poor performance leading to heart failure, whereas increased RLC phosphorylation is associated with cardiac protection from stresses. Although information is limited, RLC phosphorylation appears compromised in human heart failure which is consistent with data from mouse studies. The extent of cardiac RLC phosphorylation is determined by the balanced activities of cardiac myosin light chain kinases and phosphatases, the regulatory mechanisms of which are now emerging. This review thusly focuses on kinases that may participate in phosphorylating RLC to make the substrate for cardiac myosin light chain phosphatases, in addition to providing perspectives on the family of myosin light chain phosphatases and involved signaling mechanisms. Because biochemical and physiological information about cardiac myosin light chain phosphatase is sparse, such studies represent an emerging area of investigation in health and disease.
Collapse
Affiliation(s)
- Audrey N Chang
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Kristine E Kamm
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - James T Stull
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
43
|
Abstract
Hypertensive cardiac remodeling is characterized by left ventricular hypertrophy and interstitial fibrosis, which can lead to heart failure with preserved ejection fraction. The Rho-associated coiled-coil containing kinases (ROCKs) are members of the serine/threonine protein kinase family, which mediates the downstream effects of the small GTP-binding protein RhoA. There are 2 isoforms: ROCK1 and ROCK2. They have different functions in different types of cells and tissues. There is growing evidence that ROCKs contribute to the development of cardiovascular diseases, including cardiac fibrosis, hypertrophy, and subsequent heart failure. Recent experimental studies using ROCK inhibitors, such as fasudil, have shown the benefits of ROCK inhibition in cardiac remodeling. Mice lacking each ROCK isoform also exhibit reduced myocardial fibrosis in a variety of pathological models of cardiac remodeling. Indeed, clinical studies with fasudil have suggested that ROCKs could be potential novel therapeutic targets for cardiovascular diseases. In this review, we summarize the current understanding of the roles of ROCKs in the development of cardiac fibrosis and hypertrophy and discuss their therapeutic potential for deleterious cardiac remodeling. (Circ J 2016; 80: 1491-1498).
Collapse
Affiliation(s)
- Toru Shimizu
- Section of Cardiology, Department of Medicine, University of Chicago
| | | |
Collapse
|
44
|
Characterization of isoform expression and subcellular distribution of MYPT1 in intestinal epithelial cells. Gene 2016; 588:1-6. [PMID: 27129938 DOI: 10.1016/j.gene.2016.04.048] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Revised: 04/07/2016] [Accepted: 04/23/2016] [Indexed: 11/21/2022]
Abstract
The regulation of intestinal epithelial permeability requires phosphorylation of myosin regulatory light chain (MLC). The phosphorylation status of MLC is regulated by myosin light chain phosphatase (MLCP) activities. The activity of the catalytic subunit of MLCP (PP1cδ) toward MLC depends on its regulatory subunit (MYPT1). In this study, we revealed the presence of two MYPT1 isoforms, full length and variant 2 in human intestinal (Caco-2) epithelial cells and isolated intestinal epithelial cells (IECs) from mice. In confluent Caco-2 cells, MYPT1 was distributed at cell-cell contacts and colocalized with F-actin. These results suggest that MYPT1 isoforms are expressed in intestinal epithelial cells and MYPT1 may be involved in the regulation of intestinal epithelial barrier function.
Collapse
|
45
|
Liu B, Lee YC, Alwaal A, Wang G, Banie L, Lin CS, Lin G, Lue TF. Carbachol-induced signaling through Thr696-phosphorylation of myosin phosphatase-targeting subunit 1 (MYPT1) in rat bladder smooth muscle cells. Int Urol Nephrol 2016; 48:1237-1242. [PMID: 27118568 DOI: 10.1007/s11255-016-1303-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Accepted: 04/19/2016] [Indexed: 12/18/2022]
Abstract
PURPOSE Lines of evidence suggest that Rho-associated protein kinase (ROCK)-mediated myosin phosphatase-targeting subunit 1 (MYPT1) phosphorylation plays a central role in smooth muscle contraction. However, the physiological significance of MYPT1 phosphorylation at Thr696 catalyzed by ROCK in bladder smooth muscle remains controversial. We attempt to directly observe the quantitative protein expression of Rho A/ROCK and phosphorylation of MYPT1 at Thr696 after carbachol administration in rat bladder smooth muscle cells (RBMSCs). MATERIALS AND METHODS Primary cultured smooth muscle cells were obtained from rat bladders. The effects of both concentration and time-course induced by the muscarinic agonist carbachol were investigated by assessing the expression of Rho A/ROCK and MYPT1 phosphorylation at Thr696 using Western blot. RESULTS In the dose-course studies, carbachol showed significant increase in phosphorylation of MYPT1 at Thr696 (p-MYPT1) from concentrations of 15-100 μM based on Western blot results (p < 0.05, ANOVA test). In the time-course studies, treatment of cells with 15 μM of carbachol significantly enhanced the expression of p-MYPT1 from 3 to 15 h (p < 0.05, ANOVA test) and induced the expression of Rho A from 10 to 120 min (p < 0.05, ANOVA test). CONCLUSIONS Carbachol can induce the expression of ROCK pathway, leading to MYPT1 phosphorylation at Thr696 and thereby sustained RBSMCs contraction.
Collapse
Affiliation(s)
- Benchun Liu
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, 400 Parnassus Ave., Ste A-610, San Francisco, CA, 94143-0738, USA
| | - Yung-Chin Lee
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, 400 Parnassus Ave., Ste A-610, San Francisco, CA, 94143-0738, USA
- Department of Urology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Amjad Alwaal
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, 400 Parnassus Ave., Ste A-610, San Francisco, CA, 94143-0738, USA
| | - Guifang Wang
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, 400 Parnassus Ave., Ste A-610, San Francisco, CA, 94143-0738, USA
| | - Lia Banie
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, 400 Parnassus Ave., Ste A-610, San Francisco, CA, 94143-0738, USA
| | - Ching-Shwun Lin
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, 400 Parnassus Ave., Ste A-610, San Francisco, CA, 94143-0738, USA
| | - Guiting Lin
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, 400 Parnassus Ave., Ste A-610, San Francisco, CA, 94143-0738, USA
| | - Tom F Lue
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, 400 Parnassus Ave., Ste A-610, San Francisco, CA, 94143-0738, USA.
| |
Collapse
|
46
|
Reho JJ, Kenchegowda D, Asico LD, Fisher SA. A splice variant of the myosin phosphatase regulatory subunit tunes arterial reactivity and suppresses response to salt loading. Am J Physiol Heart Circ Physiol 2016; 310:H1715-24. [PMID: 27084390 DOI: 10.1152/ajpheart.00869.2015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 04/12/2016] [Indexed: 12/21/2022]
Abstract
The cGMP activated kinase cGK1α is targeted to its substrates via leucine zipper (LZ)-mediated heterodimerization and thereby mediates vascular smooth muscle (VSM) relaxation. One target is myosin phosphatase (MP), which when activated by cGK1α results in VSM relaxation even in the presence of activating calcium. Variants of MP regulatory subunit Mypt1 are generated by alternative splicing of the 31 nt exon 24 (E24), which, by changing the reading frame, codes for isoforms that contain or lack the COOH-terminal LZ motif (E24+/LZ-; E24-/LZ+). Expression of these isoforms is vessel specific and developmentally regulated, modulates in disease, and is proposed to confer sensitivity to nitric oxide (NO)/cGMP-mediated vasorelaxation. To test this, mice underwent Tamoxifen-inducible and smooth muscle-specific knockout of E24 (E24 cKO) after weaning. Deletion of a single allele of E24 (shift to Mypt1 LZ+) enhanced vasorelaxation of first-order mesenteric arteries (MA1) to diethylamine-NONOate (DEA/NO) and to cGMP in permeabilized and calcium-clamped arteries and lowered blood pressure. There was no further effect of deletion of both E24 alleles, indicating high sensitivity to shift of Mypt1 isoforms. However, a unique property of MA1s from homozygous E24 cKOs was significantly reduced force generation to α-adrenergic activation. Furthermore 2 wk of high-salt (4% NaCl) diet increased MA1 force generation to phenylephrine in control mice, a response that was markedly suppressed in the E24 cKO homozygotes. Thus Mypt1 E24 splice variants tune arterial reactivity and could be worthy targets for lowering vascular resistance in disease states.
Collapse
Affiliation(s)
- John J Reho
- Department of Medicine, Divisions of Cardiovascular Medicine and Nephrology, University of Maryland-Baltimore, Baltimore, Maryland
| | - Doreswamy Kenchegowda
- Department of Medicine, Divisions of Cardiovascular Medicine and Nephrology, University of Maryland-Baltimore, Baltimore, Maryland
| | - Laureano D Asico
- Department of Medicine, Divisions of Cardiovascular Medicine and Nephrology, University of Maryland-Baltimore, Baltimore, Maryland
| | - Steven A Fisher
- Department of Medicine, Divisions of Cardiovascular Medicine and Nephrology, University of Maryland-Baltimore, Baltimore, Maryland
| |
Collapse
|
47
|
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
|
48
|
Perrino BA. Calcium Sensitization Mechanisms in Gastrointestinal Smooth Muscles. J Neurogastroenterol Motil 2016; 22:213-25. [PMID: 26701920 PMCID: PMC4819859 DOI: 10.5056/jnm15186] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Accepted: 12/22/2014] [Indexed: 01/05/2023] Open
Abstract
An increase in intracellular Ca2+ is the primary trigger of contraction of gastrointestinal (GI) smooth muscles. However, increasing the Ca2+ sensitivity of the myofilaments by elevating myosin light chain phosphorylation also plays an essential role. Inhibiting myosin light chain phosphatase activity with protein kinase C-potentiated phosphatase inhibitor protein-17 kDa (CPI-17) and myosin phosphatase targeting subunit 1 (MYPT1) phosphorylation is considered to be the primary mechanism underlying myofilament Ca2+ sensitization. The relative importance of Ca2+ sensitization mechanisms to the diverse patterns of GI motility is likely related to the varied functional roles of GI smooth muscles. Increases in CPI-17 and MYPT1 phosphorylation in response to agonist stimulation regulate myosin light chain phosphatase activity in phasic, tonic, and sphincteric GI smooth muscles. Recent evidence suggests that MYPT1 phosphorylation may also contribute to force generation by reorganization of the actin cytoskeleton. The mechanisms responsible for maintaining constitutive CPI-17 and MYPT1 phosphorylation in GI smooth muscles are still largely unknown. The characteristics of the cell-types comprising the neuroeffector junction lead to fundamental differences between the effects of exogenous agonists and endogenous neurotransmitters on Ca2+ sensitization mechanisms. The contribution of various cell-types within the tunica muscularis to the motor responses of GI organs to neurotransmission must be considered when determining the mechanisms by which Ca2+ sensitization pathways are activated. The signaling pathways regulating Ca2+ sensitization may provide novel therapeutic strategies for controlling GI motility. This article will provide an overview of the current understanding of the biochemical basis for the regulation of Ca2+ sensitization, while also discussing the functional importance to different smooth muscles of the GI tract.
Collapse
Affiliation(s)
- Brian A Perrino
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA
| |
Collapse
|
49
|
MacDonald JA. A tale of two threonines: myosin phosphatase inhibition and calcium sensitization of smooth muscle. J Physiol 2016; 593:487-8. [PMID: 25774393 DOI: 10.1113/jphysiol.2014.270019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
50
|
Gao N, Chang AN, He W, Chen CP, Qiao YN, Zhu M, Kamm KE, Stull JT. Physiological signalling to myosin phosphatase targeting subunit-1 phosphorylation in ileal smooth muscle. J Physiol 2016; 594:3209-25. [PMID: 26847850 DOI: 10.1113/jp271703] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 01/21/2016] [Indexed: 12/16/2022] Open
Abstract
KEY POINTS The extent of myosin regulatory light chain phosphorylation (RLC) necessary for smooth muscle contraction depends on the respective activities of Ca(2+) /calmodulin-dependent myosin light chain kinase and myosin light chain phosphatase (MLCP), which contains a regulatory subunit MYPT1 bound to the phosphatase catalytic subunit and myosin. MYPT1 showed significant constitutive T696 and T853 phosphorylation, which is predicted to inhibit MLCP activity in isolated ileal smooth muscle tissues, with additional phosphorylation upon pharmacological treatment with the muscarinic agonist carbachol. Electrical field stimulation (EFS), which releases ACh from nerves, increased force and RLC phosphorylation but not MYPT1 T696 or T853 phosphorylation. The conditional knockout of MYPT1 or the knockin mutation T853A in mice had no effect on the frequency-maximal force responses to EFS in isolated ileal tissues. Physiological RLC phosphorylation and force development in ileal smooth muscle depend on myosin light chain kinase and MLCP activities without changes in constitutive MYPT1 phosphorylation. ABSTRACT Smooth muscle contraction initiated by myosin regulatory light chain (RLC) phosphorylation is dependent on the relative activities of Ca(2+) /calmodulin-dependent myosin light chain kinase (MLCK) and myosin light chain phosphatase (MLCP). We have investigated the physiological role of the MLCP regulatory subunit MYPT1 in ileal smooth muscle in adult mice with (1) smooth muscle-specific deletion of MYPT1; (2) non-phosphorylatable MYPT1 containing a T853A knockin mutation; and (3) measurements of force and protein phosphorylation responses to cholinergic neurostimulation initiated by electric field stimulation. Isolated MYPT1-deficient tissues from MYPT1(SM-/-) mice contracted and relaxed rapidly with moderate differences in sustained responses to KCl and carbachol treatments and washouts, respectively. Similarly, measurements of regulatory proteins responsible for RLC phosphorylation during contractions also revealed moderate changes. There were no differences in contractile or RLC phosphorylation responses to carbachol between tissues from normal mice vs. MYPT1 T853A knockin mice. Quantitatively, there was substantial MYPT1 T696 and T853 phosphorylation in wild-type tissues under resting conditions, predicting a high extent of MLCP phosphatase inhibition. Reduced PP1cδ activity in MYPT1-deficient tissues may be similar to attenuated MLCP activity in wild-type tissues resulting from constitutively phosphorylated MYPT1. Electric field stimulation increased RLC phosphorylation and force development in tissues from wild-type mice without an increase in MYPT1 phosphorylation. Thus, physiological RLC phosphorylation and force development in ileal smooth muscle appear to be dependent on MLCK and MLCP activities without changes in constitutive MYPT1 phosphorylation.
Collapse
Affiliation(s)
- Ning Gao
- Department of Physiology, University of Texas Southwestern Medical Centre, Dallas, TX, USA
| | - Audrey N Chang
- Department of Physiology, University of Texas Southwestern Medical Centre, Dallas, TX, USA
| | - Weiqi He
- Model Animal Research Centre and MOE Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing, China.,Current address: Cambridge-Suda (CAM-SU) Genomic Resource Centre, Soochow University, Suzhou, China
| | - Cai-Ping Chen
- Model Animal Research Centre and MOE Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing, China
| | - Yan-Ning Qiao
- Model Animal Research Centre and MOE Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing, China
| | - Minsheng Zhu
- Model Animal Research Centre and MOE Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing, China
| | - Kristine E Kamm
- Department of Physiology, University of Texas Southwestern Medical Centre, Dallas, TX, USA
| | - James T Stull
- Department of Physiology, University of Texas Southwestern Medical Centre, Dallas, TX, USA
| |
Collapse
|