1
|
Zou X, Yuan M, Zhou W, Cai A, Cheng Y, Zhan Z, Zhang Y, Pan Z, Hu X, Zheng S, Liu T, Huang P. SOX17 Prevents Endothelial-Mesenchymal Transition of Pulmonary Arterial Endothelial Cells in Pulmonary Hypertension through Mediating TGF-β/Smad2/3 Signaling. Am J Respir Cell Mol Biol 2025; 72:364-379. [PMID: 39392679 DOI: 10.1165/rcmb.2023-0355oc] [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: 10/09/2023] [Accepted: 10/11/2024] [Indexed: 10/12/2024] Open
Abstract
Endothelial-to-mesenchymal transition (EndMT) has been reported to contribute to pulmonary vascular remodeling in patients with pulmonary hypertension (PH). Our study demonstrates that SOX17, a member of the SOX (SRY-Box) transcription factor family, plays a role in regulating pulmonary arterial homeostasis through extracellular vesicles in an autocrine and paracrine manner. However, the role of SOX17 in mediating EndMT of pulmonary arterial endothelial cells (PAECs) and its intracellular mechanisms remain unclear. Here we present evidence showing that downregulation of SOX17 expression is accompanied by significant pulmonary arterial EndMT and activation of the TGF-β/Smad2/3 signaling pathway in patients with idiopathic PH and rats with PH induced by Sugen 5416/hypoxia. In primary human PAECs, canonical TGF-β (transforming growth factor-β) signaling inhibits the expression of SOX17. Overexpression of SOX17 reverses TGF-β- and hypoxia-induced EndMT. These findings suggest that SOX17 is essential for human PAECs to undergo TGF-β-mediated EndMT. Mechanistically, our data demonstrate that SOX17 prevents TGF-β-induced EndMT by suppressing ROCK1 (Rho-associated kinase 1) expression through binding to the specific promoter region of ROCK1, thereby inhibiting MYPT1 (myosin phosphatase target subunit 1) and MLC (myosin light chain) phosphorylation. Furthermore, we show that Tie2-Cre rats with endothelial cell-specific overexpression of SOX17 are protected against Sugen/hypoxia-induced EndMT and subsequent pulmonary vascular remodeling. Consistent with the in vitro results, compared with Tie2-Cre rats treated with Sugen/hypoxia alone, rats overexpressing SOX17 exhibited reduced levels of ROCK1 as well as decreased phosphorylation levels of MYPT1 and MLC. Overall, our studies unveil a novel TGF-β/SOX17/ROCK1 pathway involved in regulating PAECs' EndMT process, and we propose the targeting of SOX17 as a potential therapeutic strategy for alleviating pulmonary vascular remodeling in PH.
Collapse
Affiliation(s)
- Xiaozhou Zou
- Center for Clinical Pharmacy, Cancer Center, and Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China
| | - Mengnan Yuan
- Center for Clinical Pharmacy, Cancer Center, and Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China
| | - Wei Zhou
- Institute of Hepatobiliary Diseases of Wuhan University, Zhongnan Hospital of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, China
| | - Anqi Cai
- Center for Clinical Pharmacy, Cancer Center, and Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China
| | - Yili Cheng
- Center for Clinical Pharmacy, Cancer Center, and Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China
| | - Zibo Zhan
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yiwen Zhang
- Center for Clinical Pharmacy, Cancer Center, and Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China
| | - Zongfu Pan
- Center for Clinical Pharmacy, Cancer Center, and Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China
| | - Xiaoping Hu
- Center for Clinical Pharmacy, Cancer Center, and Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China
| | - Shuilian Zheng
- Center for Clinical Pharmacy, Cancer Center, and Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China
| | - Ting Liu
- Department of Pharmacy, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China; and
| | - Ping Huang
- Center for Clinical Pharmacy, Cancer Center, and Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China
| |
Collapse
|
2
|
Zhang Y, Li N, Kobayashi S. Paxillin participates in the sphingosylphosphorylcholine-induced abnormal contraction of vascular smooth muscle by regulating Rho-kinase activation. Cell Commun Signal 2024; 22:58. [PMID: 38254202 PMCID: PMC10801962 DOI: 10.1186/s12964-023-01404-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/22/2023] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND The Ca2+-independent contraction of vascular smooth muscle is a leading cause of cardiovascular and cerebrovascular spasms. In the previous study, we demonstrated the involvement of Src family protein tyrosine kinase Fyn and Rho-kinase in the sphingosylphosphorylcholine (SPC)-induced abnormal and Ca2+-independent contraction of vascular smooth muscle, but the specific mechanism has not been completely clarified. METHODS Paxillin knockdown human coronary artery smooth muscle cells (CASMCs) and smooth muscle-specific paxillin knockout mice were generated by using paxillin shRNA and the tamoxifen-inducible Cre-LoxP system, respectively. CASMCs contraction was observed by time-lapse recording. The vessel contractility was measured by using a myography assay. Fyn, Rho-kinase, and myosin light chain activation were assessed by immunoprecipitation and western blotting. The paxillin expression and actin stress fibers were visualized by histological analysis and immunofluorescent staining. RESULTS The SPC-induced abnormal contraction was inhibited in paxillin knockdown CASMCs and arteries of paxillin knockout mice, indicating that paxillin is involved in this abnormal contraction. Further study showed that paxillin knockdown inhibited the SPC-induced Rho-kinase activation without affecting Fyn activation. In addition, paxillin knockdown significantly inhibited the SPC-induced actin stress fiber formation and myosin light chain phosphorylation. These results suggest that paxillin, as an upstream molecule of Rho-kinase, is involved in the SPC-induced abnormal contraction of vascular smooth muscle. CONCLUSIONS The present study demonstrated that paxillin participates in the SPC-induced abnormal vascular smooth muscle contraction by regulating Rho-kinase activation. Video Abstract.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Yamaguchi University, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan.
| | - Nan Li
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Yamaguchi University, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Sei Kobayashi
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Yamaguchi University, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan.
| |
Collapse
|
3
|
Mironova GY, Kowalewska PM, El-Lakany M, Tran CHT, Sancho M, Zechariah A, Jackson WF, Welsh DG. The conducted vasomotor response and the principles of electrical communication in resistance arteries. Physiol Rev 2024; 104:33-84. [PMID: 37410448 PMCID: PMC11918294 DOI: 10.1152/physrev.00035.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 06/26/2023] [Accepted: 07/01/2023] [Indexed: 07/07/2023] Open
Abstract
Biological tissues are fed by arterial networks whose task is to set blood flow delivery in accordance with energetic demand. Coordinating vasomotor activity among hundreds of neighboring segments is an essential process, one dependent upon electrical information spreading among smooth muscle and endothelial cells. The "conducted vasomotor response" is a functional expression of electrical spread, and it is this process that lies at the heart of this critical review. Written in a narrative format, this review first highlights historical manuscripts and then characterizes the conducted response across a range of preparations. Trends are highlighted and used to guide subsequent sections, focused on cellular foundations, biophysical underpinnings, and regulation in health and disease. Key information has been tabulated; figures reinforce grounding concepts and reveal a framework within which theoretical and experimental work can be rationalized. This summative review highlights that despite 30 years of concerted experimentation, key aspects of the conducted response remain ill defined. Of note is the need to rationalize the regulation and deterioration of conduction in pathobiological settings. New quantitative tools, along with transgenic technology, are discussed as a means of propelling this investigative field forward.
Collapse
Affiliation(s)
- Galina Yu Mironova
- Department of Physiology and Pharmacology, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Paulina M Kowalewska
- Department of Physiology and Pharmacology, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Mohammed El-Lakany
- Department of Physiology and Pharmacology, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Cam Ha T Tran
- Department of Physiology, Faculty of Medicine, University of Nevada (Reno), Reno, Nevada, United States
| | - Maria Sancho
- Department of Physiology, Faculty of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Anil Zechariah
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland and Labrador, St. John's, Newfoundland, Canada
| | - William F Jackson
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States
| | - Donald G Welsh
- Department of Physiology and Pharmacology, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
4
|
Soh JEC, Shimizu A, Sato A, Ogita H. Novel cardiovascular protective effects of RhoA signaling and its therapeutic implications. Biochem Pharmacol 2023; 218:115899. [PMID: 37907138 DOI: 10.1016/j.bcp.2023.115899] [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: 09/19/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 11/02/2023]
Abstract
Ras homolog gene family member A (RhoA) belongs to the Rho GTPase superfamily, which was first studied in cancers as one of the essential regulators controlling cellular function. RhoA has long attracted attention as a key molecule involved in cell signaling and gene transcription, through which it affects cellular processes. A series of studies have demonstrated that RhoA plays crucial roles under both physiological states and pathological conditions in cardiovascular diseases. RhoA has been identified as an important regulator in cardiac remodeling by regulating actin stress fiber dynamics and cytoskeleton formation. However, its underlying mechanisms remain poorly understood, preventing definitive conclusions being drawn about its protective role in the cardiovascular system. In this review, we outline the characteristics of RhoA and its related signaling molecules, and present an overview of RhoA classical function and the corresponding cellular responses of RhoA under physiological and pathological conditions. Overall, we provide an update on the novel signaling under RhoA in the cardiovascular system and its potential clinical and therapeutic targets in cardiovascular medicine.
Collapse
Affiliation(s)
- Joanne Ern Chi Soh
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| | - Akio Shimizu
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| | - Akira Sato
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| | - Hisakazu Ogita
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan.
| |
Collapse
|
5
|
Portela Dias J, Guedes-Martins L. Fetal Pulmonary Venous Return: From Basic Research to the Clinical Value of Doppler Assessment. Pediatr Cardiol 2023; 44:1419-1437. [PMID: 37505268 PMCID: PMC10435640 DOI: 10.1007/s00246-023-03244-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/20/2023] [Indexed: 07/29/2023]
Abstract
The fetal pulmonary circulation represents less than 25% of the fetal cardiac output. In comparison with the pulmonary arteries, studies on pulmonary veins are few and limited, and many questions remain to be answered. The literature reports that pulmonary veins play an important role in regulating vascular flow, forming an active segment of the pulmonary circulation. The development of more sophisticated ultrasonography technology has allowed the investigation of the extraparenchymal pulmonary veins and their waveform. The recognition of the pulmonary vein anatomy in echocardiography is important for the diagnosis of anomalous pulmonary venous connections, with a significant impact on prognosis. On the other hand, the identification of the normal pulmonary vein waveform seems to be a reliable way to study left heart function, with potential applicability in fetal and maternal pathology. Thus, the goal of this narrative review was to provide a clinically oriented perspective of the available literature on this topic.
Collapse
Affiliation(s)
- J Portela Dias
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313, Porto, Portugal.
- Departamento da Mulher e da Medicina Reprodutiva, Centro Materno Infantil do Norte, Centro Hospitalar e Universitário de Santo António, Largo da Maternidade Júlio Dinis 45, 4050-651, Porto, Portugal.
- Unidade de Investigação e Formação - Centro Materno Infantil do Norte, 4050-651, Porto, Portugal.
| | - L Guedes-Martins
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313, Porto, Portugal
- Departamento da Mulher e da Medicina Reprodutiva, Centro Materno Infantil do Norte, Centro Hospitalar e Universitário de Santo António, Largo da Maternidade Júlio Dinis 45, 4050-651, Porto, Portugal
- Unidade de Investigação e Formação - Centro Materno Infantil do Norte, 4050-651, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal
| |
Collapse
|
6
|
Wang X, Xu X, Zhu Q, Han Y, Zhang W. Hypoxia-induced miR-182-5p regulates vascular smooth muscle cell phenotypic switch by targeting RGS5. Cell Biol Int 2022; 46:1864-1875. [PMID: 35946384 DOI: 10.1002/cbin.11883] [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: 02/22/2022] [Accepted: 07/08/2022] [Indexed: 11/12/2022]
Abstract
In response to vascular injury or alterations in the local environment, such as hypoxia and hypertension, contractile vascular smooth muscle cells (VSMCs) are able to switch to a synthetic phenotype characterized by increased extracellular matrix synthesis with decreased expression of contractile markers. miR-182-5p has recently been reported to play a regulatory role in VSMCs proliferation. However, little is known about its target genes and related pathways in VSMCs phenotypic switch. Here, we investigated the function of miR-182-5p in VSMCs phenotypic switch. The results showed that upregulation of miR-182-5p promoted the switching of VSMCs from a contractile to a synthetic phenotype under hypoxic conditions. Mechanistically, hypoxia elevated miR-182-5p, leading to a reduction in expression of contractile markers and weakened RhoA signaling. Using bioinformatics analysis, dual-luciferase reporter assays and rescue assays, we demonstrated that miR-182-5p suppressed RhoA signaling by targeting RGS5. Collectively, the results from the present study indicated that miR-182-5p/RGS5/RhoA axis regulated hypoxia-induced VSMCs phenotypic switch.
Collapse
Affiliation(s)
- Xiaozhou Wang
- Research Center for High Altitude Medicine, Qinghai University, Xining, Qinghai, China.,Key Laboratory for High Altitude Medicine, Ministry of Education, Xining, Qinghai, China.,Key Laboratory of Application and Foundation for High Altitude Medicine in Qinghai Province, Qinghai University, Xining, Qinghai, China.,Department of Hypertension, Qinghai Cardio-Cerebrovascular Hospital, Xining, Qinghai, China
| | - Xiaolong Xu
- Department of Hypertension, Qinghai Cardio-Cerebrovascular Hospital, Xining, Qinghai, China
| | - Qinfang Zhu
- Research Center for High Altitude Medicine, Qinghai University, Xining, Qinghai, China.,Key Laboratory for High Altitude Medicine, Ministry of Education, Xining, Qinghai, China.,Key Laboratory of Application and Foundation for High Altitude Medicine in Qinghai Province, Qinghai University, Xining, Qinghai, China.,Department of Endocrinology, Qinghai Provincial People's Hospital, Xining, Qinghai, China
| | - Ying Han
- Research Center for High Altitude Medicine, Qinghai University, Xining, Qinghai, China.,Key Laboratory for High Altitude Medicine, Ministry of Education, Xining, Qinghai, China.,Key Laboratory of Application and Foundation for High Altitude Medicine in Qinghai Province, Qinghai University, Xining, Qinghai, China
| | - Wei Zhang
- Research Center for High Altitude Medicine, Qinghai University, Xining, Qinghai, China.,Key Laboratory for High Altitude Medicine, Ministry of Education, Xining, Qinghai, China.,Key Laboratory of Application and Foundation for High Altitude Medicine in Qinghai Province, Qinghai University, Xining, Qinghai, China
| |
Collapse
|
7
|
Kubota S, Yamamoto Y, Kimura K. A Chinese Medicine, Tokishakuyakusan, Increases Bovine Oviductal Tonus <i>via</i> G Protein-Coupled Estrogen Receptor 1. Biol Pharm Bull 2022; 45:1133-1141. [DOI: 10.1248/bpb.b22-00201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Sayaka Kubota
- Department of Animal Production Science, Graduate School of Environmental and Life Science, Okayama University
| | - Yuki Yamamoto
- Department of Animal Production Science, Graduate School of Environmental and Life Science, Okayama University
| | - Koji Kimura
- Department of Animal Production Science, Graduate School of Environmental and Life Science, Okayama University
| |
Collapse
|
8
|
Effect of GNE Mutations on Cytoskeletal Network Proteins: Potential Gateway to Understand Pathomechanism of GNEM. Neuromolecular Med 2022; 24:452-468. [PMID: 35503500 DOI: 10.1007/s12017-022-08711-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 04/09/2022] [Indexed: 12/27/2022]
Abstract
GNE myopathy is an inherited neuromuscular disorder caused by mutations in GNE (UDP-N-acetylglucosamine 2-epimerase/N-acetyl mannosamine kinase) gene catalyzing the sialic acid biosynthesis pathway. The characteristic features include muscle weakness in upper and lower extremities, skeletal muscle wasting, and rimmed vacuole formation. More than 200 GNE mutations in either epimerase or kinase domain have been reported worldwide. In Indian subcontinent, several GNE mutations have been recently identified with unknown functional correlation. Alternate role of GNE in various cellular processes such as cell adhesion, migration, apoptosis, protein aggregation, and cytoskeletal organization have been proposed in recent studies. We aim to understand and compare the effect of various GNE mutations from Indian origin on regulation of the cytoskeletal network. In particular, F-actin dynamics was determined quantitatively by determining F/G-actin ratios in immunoblots for specific proteins. The extent of F-actin polymerization was visualized by immunostaining with Phalloidin using confocal microscopy. The proteins regulating F-actin dynamics such as RhoA, cofilin, Arp2, and alpha-actinin were studied in various GNE mutants. The altered level of cytoskeletal organization network proteins affected cell migration of GNE mutant proteins as measured by wound healing assay. The functional comparison of GNE mutations will help in better understanding of the genotypic severity of the disease in the Indian population. Our study offers a potential for identification of therapeutic molecules regulating actin dynamics in GNE specific mutations.
Collapse
|
9
|
Li N, Zhang Y, Morita T, Kishi H, Kobayashi S. Inhibitory mechanism of tangeretin, a citrus flavone on the sphingosylphosphorylcholine (SPC)-induced vascular smooth muscle contraction. J Pharmacol Sci 2022; 149:189-197. [DOI: 10.1016/j.jphs.2022.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/14/2022] [Accepted: 05/06/2022] [Indexed: 10/18/2022] Open
|
10
|
El-Sayed SS, Shahin RM, Fahmy A, Elshazly SM. Quercetin ameliorated remote myocardial injury induced by renal ischemia/reperfusion in rats: Role of Rho-kinase and hydrogen sulfide. Life Sci 2021; 287:120144. [PMID: 34785193 DOI: 10.1016/j.lfs.2021.120144] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 10/30/2021] [Accepted: 11/09/2021] [Indexed: 10/24/2022]
Abstract
AIMS This study was designated to investigate the means through which quercetin confers its cardioprotective action against remote cardiomyopathy elicited by renal ischemia/reperfusion (I/R). Potential involvement of hydrogen sulfide (H2S) and its related mechanisms were accentuated herein. MAIN METHODS In anesthetized male Wistar rats, renal I/R was induced by bilateral renal pedicles occlusion for 30 min (ischemia) followed by 24 h reperfusion. Quercetin (50 mg/kg, gavage) was administered at 5 h post reperfusion initiation and 2 h before euthanasia. Cystathionine β-synthase (CBS) inhibitor, amino-oxyacetic acid (AOAA; 10 mg/kg, i.p) was given 30 min prior to each quercetin dose. KEY FINDINGS Quercetin reversed renal I/R induced derangements; as quercetin administration improved renal function and reversed I/R induced histopathological changes in both myocardium and kidney. Further, quercetin enhanced renal CBS content/activity, while mitigated myocardial cystathionine ɤ-lyase (CSE) content/activity as well as myocardial H2S. On the other hand, quercetin augmented myocardial nitric oxide (NO), nuclear factor erythroid 2-related factor 2 (Nrf2) and its nuclear trasnslocation, glutamate cysteine ligase (GCL), reduced glutathione (GSH) and peroxiredoxin-2 (Prx2), while further reduced lipid peroxidation measured as malondialdehyde (MDA) as well as nuclear factor-kappa B (NF-κB), caspase-3 content and activity, and Rho-kinase activity. SIGNIFICANCE Cardioprotective effects of quercetin may be mediated through regulation of Rho-kinase pathway and H2S production.
Collapse
Affiliation(s)
- Shaimaa S El-Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt.
| | - Rania M Shahin
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt.
| | - Ahmed Fahmy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt.
| | - Shimaa M Elshazly
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt.
| |
Collapse
|
11
|
Devi SS, Yadav R, Arya R. Altered Actin Dynamics in Cell Migration of GNE Mutant Cells. Front Cell Dev Biol 2021; 9:603742. [PMID: 33816461 PMCID: PMC8012676 DOI: 10.3389/fcell.2021.603742] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 02/09/2021] [Indexed: 11/27/2022] Open
Abstract
Cell migration is an essential cellular process that requires coordination of cytoskeletal dynamics, reorganization, and signal transduction. The actin cytoskeleton is central in maintaining the cellular structure as well as regulating the mechanisms of cell motility. Glycosylation, particularly sialylation of cell surface proteins like integrins, regulates signal transduction from the extracellular matrix to the cytoskeletal network. The activation of integrin by extracellular cues leads to recruitment of different focal adhesion complex proteins (Src, FAK, paxillin, etc.) and activates the signal including Rho GTPases for the regulation of actin assembly and disassembly. During cell migration, the assembly and disassembly of actin filament provides the essential force for the cell to move. Abnormal sialylation can lead to actin signaling dysfunction leading to aberrant cell migration, one of the main characteristics of cancer and myopathies. In the present study, we have reported altered F-actin to G-actin ratios in GNE mutated cells. These cells exhibit pathologically relevant mutations of GNE (UDP N-acetylneuraminic 2-epimerase/N-acetylmannosamine kinase), a key sialic acid biosynthetic enzyme. It was found that GNE neither affects the actin polymerization nor binds directly to actin. However, mutation in GNE resulted in increased binding of α-actinin to actin filaments. Further, through confocal imaging, GNE was found to be localized in focal adhesion complex along with paxillin. We further elucidated that mutation in GNE resulted in upregulation of RhoA protein and Cofilin activity is downregulated, which could be rescued with Rhosin and chlorogenic acid, respectively. Lastly, mutant in GNE reduced cell migration as implicated from wound healing assay. Our study indicates that molecules altering Cofilin function could significantly revert the cell migration defect due to GNE mutation in sialic acid-deficient cells. We propose cytoskeletal proteins to be alternate drug targets for disorders associated with GNE such as GNE myopathy.
Collapse
Affiliation(s)
| | - Rashmi Yadav
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Ranjana Arya
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
12
|
de Oliveira AA, Priviero F, Tostes RC, Webb RC, Nunes KP. Dissecting the interaction between HSP70 and vascular contraction: role of [Formula: see text] handling mechanisms. Sci Rep 2021; 11:1420. [PMID: 33446873 PMCID: PMC7809064 DOI: 10.1038/s41598-021-80966-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 12/09/2020] [Indexed: 01/01/2023] Open
Abstract
Heat-shock protein 70 (HSP70) is a ubiquitously expressed molecular chaperone with various biological functions. Recently, we demonstrated that HSP70 is key for adequate vascular reactivity. However, the specific mechanisms targeted by HSP70 to assist in this process remain elusive. Since there is a wealth of evidence connecting HSP70 to calcium ([Formula: see text]), a master regulator of contraction, we designed this study to investigate whether blockade of HSP70 disrupts vascular contraction via impairment of [Formula: see text] handling mechanisms. We performed functional studies in aortas isolated from male Sprague Dawley rats in the presence or absence of exogenous [Formula: see text], and we determined the effects of VER155008, an inhibitor of HSP70, on [Formula: see text] handling as well as key mechanisms that regulate vascular contraction. Changes in the intracellular concentration of [Formula: see text] were measured with a biochemical assay kit. We report that blockade of HSP70 leads to [Formula: see text] mishandling in aorta stimulated with phenylephrine, decreasing both phasic and tonic contractions. Importantly, in [Formula: see text] free Krebs' solution, inhibition of HSP70 only reduced the [Formula: see text] of the phasic contraction if the protein was blocked before IP3r-mediated [Formula: see text] release, suggesting that HSP70 has a positive effect towards this receptor. Corroborating this statement, VER155008 did not potentiate an IP3r inhibitor's outcomes, even with partial blockade. In another set of experiments, the inhibition of HSP70 attenuated the amplitude of the tonic contraction independently of the moment VER155008 was added to the chamber (i.e., whether it was before or after IP3r-mediated phasic contraction). More compelling, following re-addition of [Formula: see text], VER155008 amplified the inhibitory effects of a voltage-dependent [Formula: see text] channel blocker, but not of a voltage-independent [Formula: see text] channel inhibitor, indicating that HSP70 has a positive impact on the latter. Lastly, the mechanism by which HSP70 modulates vascular contraction does not involve the [Formula: see text] sensitizer protein, Rho-kinase, nor the SERCA pump, as blockade of these proteins in the presence of VER155008 almost abolished contraction. In summary, our findings shed light on the processes targeted by HSP70 during vascular contraction and open research avenues for potential new mechanisms in vascular diseases.
Collapse
Affiliation(s)
- Amanda A. de Oliveira
- Laboratory of Vascular Physiology, Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, USA
| | - Fernanda Priviero
- Department of Physiology, Augusta University, Augusta, USA
- Department of Cell Biology and Anatomy, University of South Carolina, Columbia, USA
| | - Rita C. Tostes
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - R. Clinton Webb
- Department of Cell Biology and Anatomy, University of South Carolina, Columbia, USA
| | - Kenia P. Nunes
- Laboratory of Vascular Physiology, Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, USA
| |
Collapse
|
13
|
Zou S, Shimizu T, Yamamoto M, Shimizu S, Higashi Y, Karashima T, Saito M. Age-related differences in responses to hydrogen sulfide in the bladder of spontaneously hypertensive rats. Int J Urol 2021; 28:459-465. [PMID: 33403726 DOI: 10.1111/iju.14478] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 11/29/2020] [Indexed: 12/01/2022]
Abstract
OBJECTIVES To investigate whether a response to hydrogen sulfide donors (GYY4137 and sodium hydrosulfide) and the endogenous hydrogen sulfide system (hydrogen sulfide level and expression of cysteine aminotransferase, cystathionine β-synthase, cystathionine γ-lyase, and 3-mercaptopyruvate sulfurtransferase) in the spontaneously hypertensive rat bladder differ with age, we compared the responses of hydrogen sulfide donors to micturition and bladder relaxation, and the endogenous hydrogen sulfide system in the bladder of 18-week versus 12-week-old spontaneously hypertensive rats. METHODS GYY4137 was intravesically administered and cystometry was performed in anesthetized rats. The responses of sodium hydrosulfide were evaluated in carbachol-mediated precontracted bladder strips. Bladder hydrogen sulfide levels and expression levels of each enzyme were investigated using the methylene blue method and Western blotting, respectively. RESULTS GYY4137 treatment significantly prolonged intercontraction intervals only in 12-week-old rats. Sodium hydrosulfide-induced bladder relaxation was significantly attenuated in the strips of 18-week-old rats compared with that in 12-week-old rats. In the bladder dome, significant increases in hydrogen sulfide levels and in the expression of cystathionine β-synthase, 3-mercaptopyruvate sulfurtransferase, and cysteine aminotransferase were observed in 18-week-old rats compared with 12-week-old rats. However, cystathionine γ-lyase bands were not detected in bladder tissues of either group. CONCLUSIONS Bladder relaxation induced by hydrogen sulfide may be attenuated in spontaneously hypertensive rats in an age-dependent manner.
Collapse
Affiliation(s)
- Suo Zou
- Departments of, Department of, Pharmacology, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan
| | - Takahiro Shimizu
- Departments of, Department of, Pharmacology, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan
| | - Masaki Yamamoto
- Departments of, Department of, Pharmacology, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan
| | - Shogo Shimizu
- Departments of, Department of, Pharmacology, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan
| | - Youichirou Higashi
- Departments of, Department of, Pharmacology, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan
| | - Takashi Karashima
- Department of, Urology, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan
| | - Motoaki Saito
- Departments of, Department of, Pharmacology, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan
| |
Collapse
|
14
|
Hariharan A, Weir N, Robertson C, He L, Betsholtz C, Longden TA. The Ion Channel and GPCR Toolkit of Brain Capillary Pericytes. Front Cell Neurosci 2020; 14:601324. [PMID: 33390906 PMCID: PMC7775489 DOI: 10.3389/fncel.2020.601324] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/13/2020] [Indexed: 12/14/2022] Open
Abstract
Brain pericytes reside on the abluminal surface of capillaries, and their processes cover ~90% of the length of the capillary bed. These cells were first described almost 150 years ago (Eberth, 1871; Rouget, 1873) and have been the subject of intense experimental scrutiny in recent years, but their physiological roles remain uncertain and little is known of the complement of signaling elements that they employ to carry out their functions. In this review, we synthesize functional data with single-cell RNAseq screens to explore the ion channel and G protein-coupled receptor (GPCR) toolkit of mesh and thin-strand pericytes of the brain, with the aim of providing a framework for deeper explorations of the molecular mechanisms that govern pericyte physiology. We argue that their complement of channels and receptors ideally positions capillary pericytes to play a central role in adapting blood flow to meet the challenge of satisfying neuronal energy requirements from deep within the capillary bed, by enabling dynamic regulation of their membrane potential to influence the electrical output of the cell. In particular, we outline how genetic and functional evidence suggest an important role for Gs-coupled GPCRs and ATP-sensitive potassium (KATP) channels in this context. We put forth a predictive model for long-range hyperpolarizing electrical signaling from pericytes to upstream arterioles, and detail the TRP and Ca2+ channels and Gq, Gi/o, and G12/13 signaling processes that counterbalance this. We underscore critical questions that need to be addressed to further advance our understanding of the signaling topology of capillary pericytes, and how this contributes to their physiological roles and their dysfunction in disease.
Collapse
Affiliation(s)
- Ashwini Hariharan
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Nick Weir
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Colin Robertson
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Liqun He
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Christer Betsholtz
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Department of Medicine Huddinge (MedH), Karolinska Institutet & Integrated Cardio Metabolic Centre, Huddinge, Sweden
| | - Thomas A Longden
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, United States
| |
Collapse
|
15
|
EPAC in Vascular Smooth Muscle Cells. Int J Mol Sci 2020; 21:ijms21145160. [PMID: 32708284 PMCID: PMC7404248 DOI: 10.3390/ijms21145160] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/09/2020] [Accepted: 07/19/2020] [Indexed: 02/07/2023] Open
Abstract
Vascular smooth muscle cells (VSMCs) are major components of blood vessels. They regulate physiological functions, such as vascular tone and blood flow. Under pathological conditions, VSMCs undergo a remodeling process known as phenotypic switching. During this process, VSMCs lose their contractility and acquire a synthetic phenotype, where they over-proliferate and migrate from the tunica media to the tunica interna, contributing to the occlusion of blood vessels. Since their discovery as effector proteins of cyclic adenosine 3′,5′-monophosphate (cAMP), exchange proteins activated by cAMP (EPACs) have been shown to play vital roles in a plethora of pathways in different cell systems. While extensive research to identify the role of EPAC in the vasculature has been conducted, much remains to be explored to resolve the reported discordance in EPAC’s effects. In this paper, we review the role of EPAC in VSMCs, namely its regulation of the vascular tone and phenotypic switching, with the likely involvement of reactive oxygen species (ROS) in the interplay between EPAC and its targets/effectors.
Collapse
|
16
|
Zhang W, Liu XD, Wang JW, Meng LF, Zhang YG, Wang JY. The sphingosine-1-phosphate/RhoA/Rho associated kinases/myosin light chain pathway in detrusor of female rats is down-regulated in response to ovariectomy. Chin Med J (Engl) 2020; 133:1203-1210. [PMID: 32433052 PMCID: PMC7249712 DOI: 10.1097/cm9.0000000000000767] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Dysuria is one of the main symptoms of genitourinary syndrome of menopause, which causes serious disruption to the normal life of peri-menopausal women. Studies have shown that it is related to decrease of detrusor contractile function, but the exact mechanism is still poorly understood. Previous results have suggested that the sphingosine-1-phosphate (S1P) pathway can regulate detrusor contraction, and this pathway is affected by estrogen in various tissues. However, how estrogen affects this pathway in the detrusor has not been investigated. In this study, we detected changes of the S1P/RhoA/Rho associated kinases (ROCK)/myosin light chain (MLC) pathway in the detrusor of ovariectomized rats in order to explore the underlying mechanism of dysuria during peri-menopause. METHODS Thirty-six female Sprague-Dawley rats were randomly divided into SHAM (sham operation), OVX (ovariectomy), and E groups (ovariectomy + estrogen), with 12 rats in each group. We obtained bladder detrusor tissues from each group and examined the mRNA and protein levels of the major components of the S1P/RhoA/ROCK/MLC pathway using quantitative real-time polymerase chain reaction and Western blotting, respectively. We also quantified the content of S1P in the detrusor using an enzyme linked immunosorbent assay. Finally, we compared results between the groups with one-way analysis of variance. RESULTS The components of the S1P pathway and the RhoA/ROCK/MLC pathway of the OVX group were significantly decreased, as compared with SHAM group. The percent decreases of the components in the S1P pathway were as follows: sphingosine kinase 1 (mRNA: 39%, protein: 45%) (both P < 0.05), S1P (21.73 ± 1.09 nmol/g vs. 18.86 ± 0.69 nmol/g) (P < 0.05), and S1P receptor 2/3 (S1PR2/3) (mRNA: 25%, 27%, respectively) (P < 0.05). However, the protein expression levels of S1PR2/3 and the protein and mRNA levels of SphK2 and S1PR1 did not show significant differences between groups (P > 0.05). The percent decreases of the components in the RhoA/ROCK/MLC pathway were as follows: ROCK2 (protein: 41%, mRNA: 36%) (both P < 0.05), p-MYPT1 (protein: 54%) (P < 0.05), and p-MLC20 (protein: 47%) (P < 0.05), but there were no significant differences in the mRNA and protein levels of RhoA, ROCK1, MYPT1, and MLC20 (all P > 0.05). In addition, all of the above-mentioned decreases could be reversed after estrogen supplementation (E group vs. SHAM group) (all P > 0.05). CONCLUSION In this study, we confirmed that ovariectomy is closely associated with the down-regulation of the S1P/RhoA/ROCK/MLC pathway in the rat detrusor, which may be one mechanism of dysuria caused by decreased contractile function of the female detrusor during peri-menopause.
Collapse
Affiliation(s)
- Wei Zhang
- Peking University Fifth School of Clinical Medicine, Beijing 100730, China
- Department of Urology, Beijing Hospital, National Center of Gerontology, Beijing 100730, China
| | - Xiao-Dong Liu
- Department of Urology, Beijing Hospital, National Center of Gerontology, Beijing 100730, China
| | - Jia-Wen Wang
- Department of Urology, Beijing Hospital, National Center of Gerontology, Beijing 100730, China
| | - Ling-Feng Meng
- Department of Urology, Beijing Hospital, National Center of Gerontology, Beijing 100730, China
| | - Yao-Guang Zhang
- Department of Urology, Beijing Hospital, National Center of Gerontology, Beijing 100730, China
| | - Jian-Ye Wang
- Peking University Fifth School of Clinical Medicine, Beijing 100730, China
- Department of Urology, Beijing Hospital, National Center of Gerontology, Beijing 100730, China
| |
Collapse
|
17
|
Sancho M, Welsh DG. K IR channels in the microvasculature: Regulatory properties and the lipid-hemodynamic environment. CURRENT TOPICS IN MEMBRANES 2020; 85:227-259. [PMID: 32402641 DOI: 10.1016/bs.ctm.2020.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Basal tone and perfusion control is set in cerebral arteries by the sensing of pressure and flow, key hemodynamic stimuli. These forces establish a contractile foundation within arterial networks upon which local neurovascular stimuli operate. This fundamental process is intimately tied to arterial VM and the rise in cytosolic [Ca2+] by the graded opening of voltage-operated Ca2+ channels. Arterial VM is in turn controlled by a dynamic interaction among several resident ion channels, KIR being one of particular significance. As the name suggests, KIR displays strong inward rectification, retains a small outward component, potentiated by extracellular K+ and blocked by micromolar Ba2+. Cerebrovascular KIR is unique from other K+ currents as it is present in both smooth muscle and endothelium yet lacking in classical regulatory modulation. Such observations have fostered the view that KIR is nothing more than a background conductance, activated by extracellular K+ and which passively facilitates dilation. Recent work in cell model systems has; however, identified two membrane lipids, phosphatidylinositol 4,5-bisphosphate (PIP2) and cholesterol, that interact with KIR2.x, to stabilize the channel in the preferred open or silent state, respectively. Translating this unique form of regulation, recent studies have demonstrated that specific lipid-protein interactions enable unique KIR populations to sense distinct hemodynamic stimuli and set basal tone. This review summarizes the current knowledge of vascular KIR channels and how the lipid and hemodynamic impact their activity.
Collapse
Affiliation(s)
- Maria Sancho
- Robarts Research Institute and the Department of Physiology & Pharmacology, University of Western Ontario, London, ON, Canada
| | - Donald G Welsh
- Robarts Research Institute and the Department of Physiology & Pharmacology, University of Western Ontario, London, ON, Canada.
| |
Collapse
|
18
|
Zhang M, Chang Z, Zhang P, Jing Z, Yan L, Feng J, Hu Z, Xu Q, Zhou W, Ma P, Hao Y, Zhou R. Protective effects of 18β-glycyrrhetinic acid on pulmonary arterial hypertension via regulation of Rho A/Rho kinsase pathway. Chem Biol Interact 2019; 311:108749. [DOI: 10.1016/j.cbi.2019.108749] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/26/2019] [Accepted: 07/15/2019] [Indexed: 11/28/2022]
|
19
|
Strassheim D, Gerasimovskaya E, Irwin D, Dempsey EC, Stenmark K, Karoor V. RhoGTPase in Vascular Disease. Cells 2019; 8:E551. [PMID: 31174369 PMCID: PMC6627336 DOI: 10.3390/cells8060551] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/24/2019] [Accepted: 05/27/2019] [Indexed: 12/24/2022] Open
Abstract
Ras-homologous (Rho)A/Rho-kinase pathway plays an essential role in many cellular functions, including contraction, motility, proliferation, and apoptosis, inflammation, and its excessive activity induces oxidative stress and promotes the development of cardiovascular diseases. Given its role in many physiological and pathological functions, targeting can result in adverse effects and limit its use for therapy. In this review, we have summarized the role of RhoGTPases with an emphasis on RhoA in vascular disease and its impact on endothelial, smooth muscle, and heart and lung fibroblasts. It is clear from the various studies that understanding the regulation of RhoGTPases and their regulators in physiology and pathological conditions is required for effective targeting of Rho.
Collapse
Affiliation(s)
- Derek Strassheim
- Cardiovascular and Pulmonary Research Lab, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
| | - Evgenia Gerasimovskaya
- Cardiovascular and Pulmonary Research Lab, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
- Department of Pediatrics, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
| | - David Irwin
- Cardiovascular and Pulmonary Research Lab, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
| | - Edward C Dempsey
- Cardiovascular and Pulmonary Research Lab, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
- Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045, USA.
| | - Kurt Stenmark
- Cardiovascular and Pulmonary Research Lab, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
- Department of Pediatrics, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
| | - Vijaya Karoor
- Cardiovascular and Pulmonary Research Lab, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
- Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
| |
Collapse
|
20
|
Ravarotto V, Simioni F, Sabbadin C, Pagnin E, Maiolino G, Armanini D, Calò LA. Proinflammatory/profibrotic effects of aldosterone in Gitelman's syndrome, a human model opposite to hypertension. J Endocrinol Invest 2019; 42:521-526. [PMID: 30136149 DOI: 10.1007/s40618-018-0942-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 08/09/2018] [Indexed: 12/22/2022]
Abstract
PURPOSE Aldosterone proinflammatory/profibrotic effects are mediated by the induction of mononuclear leucocytes (MNL) to express oxidative stress (OxSt)-related proteins, such as p22phox, and by the activation of RhoA/Rho kinase pathway. Gitelman's syndrome (GS), an autosomal recessive tubulopathy, is an interesting opposite model to hypertension, being characterized by hypokalemia, activation of renin-angiotensin-aldosterone system yet normo/hypotension and lack of cardiovascular-renal remodeling. We aimed to evaluate the proinflammatory/profibrotic effect of aldosterone in MNL of 6 GS patients compared with 6 healthy subjects (HS). METHODS p22phox expression and MYPT-1 phosphorylation status, a marker of RhoA/Rho kinase pathway activation, were evaluated in MNL of GS patients and HS at baseline and after incubation with aldosterone (1 × 10-8 M) alone or with canrenone (1 × 10-6 M). RESULTS At basal condition, p22phox expression was significantly higher in HS than in GS patients (1.02 ± 0.05 densitometric unit (du) vs 0.40 ± 0.1 du, respectively). Aldosterone significantly increased p22phox expression in HS and this effect was reversed by coincubation with canrenone (1.4 ± 0.05 du and 1.09 ± 0.03 du, respectively). No significant change was reported in GS after incubation of MNL with aldosterone and/or canrenone compared with basaline. Even MYPT-1 phosphorylation was significantly higher in HS compared with GS patients at basal condition (1.16 ± 0.1 du vs 0.69 ± 0.07, respectively). Aldosterone significantly increased MYPT-1 phosphorylation only in HS (1.37 ± 0.1 du vs 0.83 ± 0.12 du in GS). CONCLUSIONS GS patients seem to be protected by the OxSt status induced by aldosterone and revealed in HS. This human model could provide additional clues to highlight the proinflammatory/cardiovascular remodeling effects of aldosterone.
Collapse
Affiliation(s)
- V Ravarotto
- Department of Medicine-Nephrology, University of Padova, Via Giustiniani, 2, 35128, Padua, Italy
| | - F Simioni
- Department of Medicine-Nephrology, University of Padova, Via Giustiniani, 2, 35128, Padua, Italy
| | - C Sabbadin
- Department of Medicine-Endocrinology, University of Padova, Padua, Italy
| | - E Pagnin
- Department of Medicine-Nephrology, University of Padova, Via Giustiniani, 2, 35128, Padua, Italy
| | - G Maiolino
- Department of Medicine-Hypertension, University of Padova, Padua, Italy
| | - D Armanini
- Department of Medicine-Endocrinology, University of Padova, Padua, Italy
| | - L A Calò
- Department of Medicine-Nephrology, University of Padova, Via Giustiniani, 2, 35128, Padua, Italy.
| |
Collapse
|
21
|
Chen P, Xiao H, Huang W, Xu DQ, Guo YM, Wang X, Wang XH, DiSanto ME, Zhang XH. Testosterone regulates myosin II isoforms expression and functional activity in the rat prostate. Prostate 2018; 78:1283-1298. [PMID: 30073674 DOI: 10.1002/pros.23702] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 07/11/2018] [Indexed: 01/24/2023]
Abstract
BACKGROUND Benign prostatic hyperplasia (BPH) is mainly caused by increased prostatic smooth muscle (SM) tone and prostatic volume. At the molecular level, SM myosin II (SMM II) and non-muscle myosin II (NMM II) mediate SM tone and cell proliferation while testosterone (T) plays a permissive role in the development of BPH. AIMS The novel objective of this study was to elucidate the effects of T on the proliferation and apoptosis of rat prostatic cells and SM contractility as well as related regulatory signaling pathways. MATERIALS AND METHODS Briefly, 36 male rats were divided into three groups (sham-operated, surgically castrated, and castrated with T supplementation). In vitro organ bath studies, competitive RT-PCR, Western-blotting analysis, Masson's trichrome staining, and immunofluorescence staining were performed. RESULTS Our data showed that castration dramatically increased prostatic SM contractility and SM MHC immunostaining revealed a relatively increased SM cell numbers in the stroma. T deprivation altered prostate SMM II isoform composition with upregulation of SM-B and SM2 but downregulation of LC17a, favoring a faster more phasic-type contraction. Moreover, protein expressions of MLCK, p-MLCP, RhoB, ROCK1, and ROCK2 increased in castrated rats. Meanwhile NMM II heavy chain isoforms A, B, and C (NMMHC-A, B, and C isoforms) were altered by castration which may be linked to decreased cell proliferation and increased apoptosis. CONCLUSION Our novel data demonstrated T regulates SMM II and NMM II and their functional activities in rat prostate and T ablation not only decreases prostate size (static component) but also changes the prostatic SM tone (dynamic component).
Collapse
Affiliation(s)
- Ping Chen
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, P.R. China
| | - He Xiao
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, P.R. China
| | - Wei Huang
- Department of Urology, People's Hospital of Tuanfeng County, Hubei, China
| | - De-Qiang Xu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, P.R. China
| | - Yu-Ming Guo
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, P.R. China
| | - Xiao Wang
- Department of Urology, People's Hospital of Wuhan University, Wuhan, China
| | - Xing-Huan Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, P.R. China
| | - Michael E DiSanto
- Departments of Biomedical Sciences and Surgery, Cooper Medical School of Rowan University, Camden, New Jersey
| | - Xin-Hua Zhang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, P.R. China
| |
Collapse
|
22
|
Involvement of inhibitor kappa B kinase 2 (IKK2) in the regulation of vascular tone. J Transl Med 2018; 98:1311-1319. [PMID: 29785049 DOI: 10.1038/s41374-018-0061-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 03/22/2018] [Accepted: 03/23/2018] [Indexed: 11/09/2022] Open
Abstract
Inhibitor kappa B kinase 2 (IKK2) plays an essential role in the activation of nuclear factor kappa B (NF-kB). Recently, it has been suggested that IKK2 acts as a myosin light chain kinase (MLCK) and contributes to vasoconstriction in mouse aorta. However, the underlying mechanisms are still unknown. Therefore, we investigated whether IKK2 acts as a MLCK or regulates the activity of myosin light chain phosphatase (MLCP). Pressure myograph was used to measure vascular tone in rat mesenteric arteries. Immunofluorescence staining was performed to identify phosphorylation levels of MLC (ser19), MYPT1 (thr853 and thr696) and CPI-17 (thr38). SC-514 (IKK2 inhibitor, 50 μM) induced relaxation in the mesenteric arteries pre-contracted with 70 mM high K+ solution or U-46619 (thromboxane analog, 5 μM). The relaxation induced by SC-514 was increased in the arteries pre-contracted with U-46619 compared to arteries pre-contracted with 70 mM high K+ solution. U-46619-induced contraction was decreased by treatment of SC-514 in the presence of MLCK inhibitor, ML-7 (10 μM). In the absence of intracellular Ca2+, U-46619 still induced contraction, which was decreased by treatment of SC-514. Furthermore, phosphorylation levels of MLC (ser19) and MYPT1 (thr853) were decreased by treatment of SC-514. IKK2 is involved in the vascular contraction through regulation of MLCP activity by phosphorylating MYPT1 at thr853 in rat mesenteric arteries. These findings suggest IKK2 could be a new pharmacological target for specific therapies of various vascular diseases.
Collapse
|
23
|
Ge J, Han T, Li X, Shan L, Zhang J, Hong Y, Xia Y, Wang J, Hou M. S-adenosyl methionine regulates calcium channels and inhibits uterine smooth muscle contraction in rats with infectious premature delivery through the transient receptor protein 3/protein kinase Cβ/C-kinase-activated protein phosphatase-1 inhibitor of 17 kDa signaling pathway. Exp Ther Med 2018; 16:103-112. [PMID: 29896230 PMCID: PMC5995051 DOI: 10.3892/etm.2018.6164] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 03/09/2018] [Indexed: 12/28/2022] Open
Abstract
The aim of the present study was to investigate the effects of S-adenosyl methionine (SAMe) on infectious premature inflammatory factors and uterine contraction, and to further explore its mechanism of action via the transient receptor protein 3 (TRPC3)/protein kinase Cβ (PKCβ)/C-kinase-activated protein phosphatase-1 inhibitor of 17 kDa (CPI-17) signaling pathway, following intervention by a TRPC3 inhibitor. A rat model of premature delivery induced by lipopolysaccharide (LPS) was established. Following treatment with SAMe and inhibiting TRPC3 expression, rat serum and uterus were isolated. Hematoxylin and eosin staining was used to observe the histopathological changes in the uterus. Uterine muscle strips in vitro were selected to measure the changes in muscle tension. ELISA was utilized to measure the changes in serum inflammatory factor and oxidative stress indexes. Immunohistochemistry, western blot assay and reverse transcription-quantitative polymerase chain reaction were applied to detect calcium channel protein expression in the uterus. Western blot analysis was employed to measure the expression of TRPC3/PKCβ/CPI-17 signaling pathway-related proteins. TRPC3 was highly expressed in the uterus of rat models of premature delivery induced by LPS. Following treatment with SAMe, inflammatory cell infiltration markedly reduced in the uterus and the tension of in vitro uterine muscle strips significantly decreased. SAMe treatment suppressed inflammatory reaction and oxidative stress, and diminished L-type and T-type calcium channel protein expression. TRPC3/PKCβ/CPI-17 signaling pathway-related protein expression was also reduced. When TRPC3 expression was suppressed, the effects of SAMe against inflammation and oxidative stress were diminished. TRPC3/PKCβ/CPI-17 signaling pathway-related protein expression significantly increased. SAMe was able to reduce inflammatory reaction and oxidative stress in the uterus of rat model of infectious premature delivery induced by LPS, prolong delivery time, reduce the mortality rate of offspring rats, and serve a therapeutic role. This effect is likely achieved via the regulation of uterine contractions and childbirth through the TRPC3/PKCβ/CPI-17 signaling pathway.
Collapse
Affiliation(s)
- Jing Ge
- Department of Maternity, General Hospital of Shenyang Military Area Command, Shenyang, Liaoning 110016, P.R. China
| | - Tao Han
- Department of Oncology, General Hospital of Shenyang Military Area Command, Shenyang, Liaoning 110016, P.R. China
| | - Xiaoqiu Li
- Department of Neurology, General Hospital of Shenyang Military Area Command, Shenyang, Liaoning 110016, P.R. China
| | - Lili Shan
- Department of Maternity, General Hospital of Shenyang Military Area Command, Shenyang, Liaoning 110016, P.R. China
| | - Jinhuan Zhang
- Department of Maternity, General Hospital of Shenyang Military Area Command, Shenyang, Liaoning 110016, P.R. China
| | - Yan Hong
- Department of Maternity, General Hospital of Shenyang Military Area Command, Shenyang, Liaoning 110016, P.R. China
| | - Yanqiu Xia
- Department of Maternity, General Hospital of Shenyang Military Area Command, Shenyang, Liaoning 110016, P.R. China
| | - Jun Wang
- Department of Maternity, General Hospital of Shenyang Military Area Command, Shenyang, Liaoning 110016, P.R. China
| | - Mingxiao Hou
- Department of Cardiothoracic Surgery, General Hospital of Shenyang Military Area Command, Shenyang, Liaoning 110016, P.R. China
| |
Collapse
|
24
|
Elrashidy RA, Zhang J, Liu G. Long-term consumption of Western diet contributes to endothelial dysfunction and aortic remodeling in rats: Implication of Rho-kinase signaling. Clin Exp Hypertens 2018; 41:174-180. [DOI: 10.1080/10641963.2018.1462375] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Rania A. Elrashidy
- Department of Surgery, MetroHealth Medical Center, Case Western Reserve University, Cleveland, OH, USA
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Jing Zhang
- Department of Hyperbaric Oxygen, Capital Medical University Beijing Chao-Yang Hospital, Beijing, China
| | - Guiming Liu
- Department of Surgery, MetroHealth Medical Center, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
25
|
Chen P, Yin J, Guo YM, Xiao H, Wang XH, DiSanto ME, Zhang XH. The expression and functional activities of smooth muscle myosin and non-muscle myosin isoforms in rat prostate. J Cell Mol Med 2017; 22:576-588. [PMID: 28990332 PMCID: PMC5742693 DOI: 10.1111/jcmm.13345] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 07/10/2017] [Indexed: 11/30/2022] Open
Abstract
Benign prostatic hyperplasia (BPH) is mainly caused by increased prostatic smooth muscle (SM) tone and volume. SM myosin (SMM) and non-muscle myosin (NMM) play important roles in mediating SM tone and cell proliferation, but these molecules have been less studied in the prostate. Rat prostate and cultured primary human prostate SM and epithelial cells were utilized. In vitro organ bath studies were performed to explore contractility of rat prostate. SMM isoforms, including SM myosin heavy chain (MHC) isoforms (SM1/2 and SM-A/B) and myosin light chain 17 isoforms (LC17a/b ), and isoform ratios were determined via competitive RT-PCR. SM MHC and NM MHC isoforms (NMMHC-A, NMMHC-B and NMMHC-C) were further analysed via Western blotting and immunofluorescence microscopy. Prostatic SM generated significant force induced by phenylephrine with an intermediate tonicity between phasic bladder and tonic aorta type contractility. Correlating with this kind of intermediate tonicity, rat prostate mainly expressed LC17a and SM1 but with relatively equal expression of SM-A/SM-B at the mRNA level. Meanwhile, isoforms of NMMHC-A, B, C were also abundantly present in rat prostate with SMM present only in the stroma, while NMMHC-A, B, C were present both in the stroma and endothelial. Additionally, the SMM selective inhibitor blebbistatin could potently relax phenylephrine pre-contracted prostate SM. In conclusion, our novel data demonstrated the expression and functional activities of SMM and NMM isoforms in the rat prostate. It is suggested that the isoforms of SMM and NMM could play important roles in BPH development and bladder outlet obstruction.
Collapse
Affiliation(s)
- Ping Chen
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jing Yin
- Department of Rehabilitation, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yu-Ming Guo
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - He Xiao
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xing-Huan Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Michael E DiSanto
- Department of Surgery and Biomedical Sciences of Cooper Medical School of Rowan University, Camden, NJ, USA
| | - Xin-Hua Zhang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
26
|
Choo SH, Lee SW, Chae MR, Kang SJ, Sung HH, Han DH, Chun JN, Park JK, Kim CY, Kim HK, So I. Effects of eupatilin on the contractility of corpus cavernosal smooth muscle through nitric oxide-independent pathways. Andrology 2017; 5:1016-1022. [PMID: 28719725 DOI: 10.1111/andr.12397] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 05/26/2017] [Accepted: 06/09/2017] [Indexed: 12/19/2022]
Abstract
Eupatilin (5,7-dihydroxy-3,4,6-trimethoxyflavone) is one of the main compounds present in Artemisia species. Eupatilin has both antioxidative and anti-inflammatory properties and a relaxation effect on vascular contraction regardless of endothelial function. We evaluated the relaxant effects of eupatilin on the corpus cavernosum (CC) of rabbits and the underlying mechanisms of its activity in human corpus cavernosum smooth muscle (CCSM) cells. Isolated rabbit CC strips were mounted in an organ bath system. A conventional whole-cell patch clamp technique was used to measure activation of calcium-sensitive K+ -channel currents in human CCSM cells. The relaxation effect of eupatilin was evaluated by cumulative addition (10-5 m ~ 3 × 10-4 m) to CC strips precontracted with 10-5 m phenylephrine. Western blotting analysis was performed to measure myosin phosphatase targeting subunit 1 (MYPT1) and protein kinase C-potentiated inhibitory protein for heterotrimeric myosin light chain phosphatase of 17-kDa (CPI-17) expression and to evaluate the effect of eupatilin on the RhoA/Rho-kinase pathway. Eupatilin effectively relaxed the phenylephrine-induced tone in the rabbit CC strips in a concentration-dependent manner with an estimated EC50 value of 1.2 ± 1.6 × 10-4 m (n = 8, p < 0.05). Iberiotoxin and tetraethylammonium significantly reduced the relaxation effect (n = 8, p < 0.001 and p = 0.003, respectively). Removal of the endothelium or the presence of L-NAME or indomethacin did not affect the relaxation effect of eupatilin. In CCSM cells, the extracellular application of eupatilin 10-4 m significantly increased the outward currents, and the eupatilin-stimulated currents were significantly attenuated by treatment with 10-7 m iberiotoxin (n = 13, p < 0.05). Eupatilin reduced the phosphorylation level of MYPT1 at Thr853 of MLCP and CPI-17 at Thr38. Eupatilin-induced relaxation of the CCSM cells via NO-independent pathways. The relaxation effects of eupatilin on CCSM cells were partially due to activation of BKCa channels and inhibition of RhoA/Rho-kinase.
Collapse
Affiliation(s)
- S H Choo
- Department of Urology, Ajou University School of Medicine, Suwon, Korea
| | - S W Lee
- Department of Urology, Samsung Medical Center, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - M R Chae
- Department of Urology, Samsung Medical Center, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - S J Kang
- Department of Urology, Samsung Medical Center, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - H H Sung
- Department of Urology, Samsung Medical Center, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - D H Han
- Department of Urology, Samsung Medical Center, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - J N Chun
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - J K Park
- Department of Urology, Institute for Medical Sciences, Medical School, Chonbuk National University, Research Institute and Clinical Trial Center of Medical Device of Chonbuk National University Hospital, Jeonju, Korea
| | - C Y Kim
- College of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Korea
| | - H K Kim
- College of Pharmacy, Kyungsung University, Busan, Korea
| | - I So
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
27
|
Shintani N, Ishiyama T, Kotoda M, Asano N, Sessler DI, Matsukawa T. The effects of Y-27632 on pial microvessels during global brain ischemia and reperfusion in rabbits. BMC Anesthesiol 2017; 17:38. [PMID: 28270098 PMCID: PMC5341179 DOI: 10.1186/s12871-017-0331-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 02/24/2017] [Indexed: 11/29/2022] Open
Abstract
Background Global brain ischemia-reperfusion during propofol anesthesia provokes persistent cerebral pial constriction. Constriction is likely mediated by Rho-kinase. Cerebral vasoconstriction possibly exacerbates ischemic brain injury. Because Y-27632 is a potent Rho-kinase inhibitor, it should be necessary to evaluate its effects on cerebral pial vessels during ischemia—reperfusion period. We therefore tested the hypotheses that Y-27632 dilates cerebral pial arterioles after the ischemia-reperfusion injury, and evaluated the time-course of cerebral pial arteriolar status after the ischemia-reperfusion. Methods Japanese white rabbits were anesthetized with propofol, and a closed cranial window inserted over the left hemisphere. Global brain ischemia was produced by clamping the brachiocephalic, left common carotid, and left subclavian arteries for 15 min. Rabbits were assigned to cranial window perfusion with: (1) artificial cerebrospinal fluid (Control group, n = 7); (2) topical infusion of Y-27632 10−6 mol · L−1 for 30 min before the initiation of global brain ischemia (Pre group, n = 7); (3) topical infusion of Y-27632 10−6 mol · L−1 starting 30 min before ischemia and continuing throughout the study period (Continuous group, n = 7); and, (4) topical infusion of Y-27632 10−6 mol · L−1 starting 10 min after the ischemia and continuing until the end of the study (Post group, n = 7). Cerebral pial arterial and venule diameters were recorded 30 min before ischemia, just before arterial clamping, 10 min after clamping, and 5, 10, 20, 40, 60, 80, 100, and 120 min after unclamping. Results Mean arterial blood pressure and blood glucose concentration increased significantly after global brain ischemia except in the Continuous group. In the Pre and Continuous groups, topical application of Y-27632 produced dilation of large (mean 18–19%) and small (mean; 25–29%) pial arteries, without apparent effect on venules. Compared with the Control and Pre groups, arterioles were significantly dilated during the reperfusion period in the Continuous and Post groups (mean at 120 min: 5–8% in large arterioles and 11–12% in small arterioles). Conclusions Y-27632 dilated cerebral pial arterioles during reperfusion. Y-27632 may enhance recovery from ischemia by preventing arteriolar vasoconstriction during reperfusion.
Collapse
Affiliation(s)
- Noriyuki Shintani
- Surgical Center, University of Yamanashi Hospital, 1110 Shimokato, Chuo, 409-3898, Yamanashi, Japan.
| | - Tadahiko Ishiyama
- Surgical Center, University of Yamanashi Hospital, 1110 Shimokato, Chuo, 409-3898, Yamanashi, Japan
| | - Masakazu Kotoda
- Department of Anesthesiology, Faculty of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, 409-3898, Yamanashi, Japan
| | - Nobumasa Asano
- Department of Anesthesiology, Faculty of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, 409-3898, Yamanashi, Japan
| | - Daniel I Sessler
- Department of Outcomes Research, Cleveland Clinic, Anesthesiology Institute, Cleveland, OH, USA
| | - Takashi Matsukawa
- Department of Anesthesiology, Faculty of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, 409-3898, Yamanashi, Japan
| |
Collapse
|
28
|
Ardanaz N, Pagano PJ. Hydrogen Peroxide as a Paracrine Vascular Mediator: Regulation and Signaling Leading to Dysfunction. Exp Biol Med (Maywood) 2016; 231:237-51. [PMID: 16514169 DOI: 10.1177/153537020623100302] [Citation(s) in RCA: 168] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Numerous studies have demonstrated the ability of a variety of vascular cells, including endothelial cells, smooth muscle cells, and fibroblasts, to produce reactive oxygen species (ROS). Until recently, major emphasis was placed on the production of superoxide anion (O2–) in the vasculature as a result of its ability to directly attenuate the biological activity of endothelium-derived nitric oxide (NO). The short half-life and radius of diffusion of O2– drastically limit the role of this ROS as an important paracrine hormone in vascular biology. On the contrary, in recent years, the O2– metabolite hydrogen peroxide (H2O2) has increasingly been viewed as an important cellular signaling agent in its own right, capable of modulating both contractile and growth-promoting pathways with more far-reaching effects. In this review, we will assess the vascular production of H2O2, its regulation by endogenous scavenger systems, and its ability to activate a variety of vascular signaling pathways, thereby leading to vascular contraction and growth. This discussion will include the ability of H2O2 to (i) Initiate calcium flux as well as (ii) stimulate pathways leading to sensitization of contractile elements to calcium. The latter involves a variety of protein kinases that have also been strongly implicated in vascular hypertrophy. Previous Intensive study has emphasized the ability of NADPH oxidase-derived O2– and H2O2 to activate these pathways in cultured smooth muscle cells. However, growing evidence indicates a considerably more complex array of unique oxidase systems in the endothelium, media, and adventitia that appear to participate in these deleterious effects in a sequential and temporal manner. Taken together, these findings seem consistent with a paracrine effect of H2O2 across the vascular wall.
Collapse
Affiliation(s)
- Noelia Ardanaz
- Hypertension and Vascular Research Division, RM 7044, E&R Building, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI 48202-2689, USA
| | | |
Collapse
|
29
|
Krishnamoorthy G, Reimann K, Wangemann P. Ryanodine-induced vasoconstriction of the gerbil spiral modiolar artery depends on the Ca 2+ sensitivity but not on Ca 2+ sparks or BK channels. BMC PHYSIOLOGY 2016; 16:6. [PMID: 27806708 PMCID: PMC5093982 DOI: 10.1186/s12899-016-0026-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 10/13/2016] [Indexed: 01/26/2023]
Abstract
Background In many vascular smooth muscle cells (SMCs), ryanodine receptor-mediated Ca2+ sparks activate large-conductance Ca2+-activated K+ (BK) channels leading to lowered SMC [Ca2+]i and vasodilation. Here we investigated whether Ca2+ sparks regulate SMC global [Ca2+]i and diameter in the spiral modiolar artery (SMA) by activating BK channels. Methods SMAs were isolated from adult female gerbils, loaded with the Ca2+-sensitive flourescent dye fluo-4 and pressurized using a concentric double-pipette system. Ca2+ signals and vascular diameter changes were recorded using a laser-scanning confocal imaging system. Effects of various pharmacological agents on Ca2+ signals and vascular diameter were analyzed. Results Ca2+ sparks and waves were observed in pressurized SMAs. Inhibition of Ca2+ sparks with ryanodine increased global Ca2+ and constricted SMA at 40 cmH2O but inhibition of Ca2+ sparks with tetracaine or inhibition of BK channels with iberiotoxin at 40 cmH2O did not produce a similar effect. The ryanodine-induced vasoconstriction observed at 40 cmH2O was abolished at 60 cmH2O, consistent with a greater Ca2+-sensitivity of constriction at 40 cmH2O than at 60 cmH2O. When the Ca2+-sensitivity of the SMA was increased by prior application of 1 nM endothelin-1, ryanodine induced a robust vasoconstriction at 60 cmH2O. Conclusions The results suggest that Ca2+ sparks, while present, do not regulate vascular diameter in the SMA by activating BK channels and that the regulation of vascular diameter in the SMA is determined by the Ca2+-sensitivity of constriction.
Collapse
Affiliation(s)
- Gayathri Krishnamoorthy
- Anatomy & Physiology Department, Cell Physiology Laboratory, Kansas State University, 228 Coles Hall, Manhattan, Kansas, 66506-5802, USA
| | - Katrin Reimann
- Anatomy & Physiology Department, Cell Physiology Laboratory, Kansas State University, 228 Coles Hall, Manhattan, Kansas, 66506-5802, USA.,Department of Otolaryngology-Head and Neck Surgery, Tübingen Hearing Research Centre, and Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Philine Wangemann
- Anatomy & Physiology Department, Cell Physiology Laboratory, Kansas State University, 228 Coles Hall, Manhattan, Kansas, 66506-5802, USA.
| |
Collapse
|
30
|
Dai ZK, Kao CL, Hsieh SL, Chen IJ, Wu BN. Restoration of uridine 5'-triphosphate-suppressed delayed rectifying K + currents by an NO activator KMUP-1 involves RhoA/Rho kinase signaling in pulmonary artery smooth muscle cells. Kaohsiung J Med Sci 2016; 32:607-613. [PMID: 27914611 DOI: 10.1016/j.kjms.2016.09.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 09/19/2016] [Accepted: 09/29/2016] [Indexed: 12/01/2022] Open
Abstract
We have demonstrated that KMUP-1 (7-[2-[4-(2-chlorobenzene)piperazinyl]ethyl]-1,3-dimethylxanthine) blunts monocrotaline-induced pulmonary arterial hypertension by altering Ca2+ sensitivity, K+-channel function, endothelial nitric oxide synthase activity, and RhoA/Rho kinase (ROCK) expression. This study further investigated whether KMUP-1 impedes uridine 5'-triphosphate (UTP)-inhibited delayed rectifying K+ (KDR) current in rat pulmonary arteries involved the RhoA/ROCK signaling. Pulmonary artery smooth muscle cells (PASMCs) were enzymatically dissociated from rat pulmonary arteries. KMUP-1 (30μM) attenuated UTP (30μM)-mediated membrane depolarization and abolished UTP-enhanced cytosolic Ca2+ concentration. Whole-cell patch-clamp electrophysiology was used to monitor KDR currents. A voltage-dependent KDR current was isolated and shown to consist of a 4-aminopyridine (5mM)-sensitive component and an insensitive component. The 4-aminopyridine sensitive KDR current was suppressed by UTP (30μM). The ROCK inhibitor Y27632 (30μM) abolished the ability of UTP to inhibit the KDR current. Like Y27632, KMUP-1 (30μM) similarly abolished UTP-inhibited KDR currents. Superfused protein kinase A and protein kinase G inhibitors (KT5720, 300nM and KT5823, 300nM) did not affect UTP-inhibited KDR currents, but the currents were restored by adding KMUP-1 (30μM) to the superfusate. KMUP-1 reversal of KDR current inhibition by UTP predominantly involves the ROCK inhibition. The results indicate that the RhoA/ROCK signaling pathway plays a key role in eliciting PASMCs depolarization caused by UTP, which would result in pulmonary artery constriction. KMUP-1 blocks UTP-mediated PASMCs depolarization, suggesting that it would prevent abnormal pulmonary vasoconstriction.
Collapse
Affiliation(s)
- Zen-Kong Dai
- Department of Pediatrics, Division of Pediatric Pulmonology and Cardiology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chang-Ling Kao
- Department of Pharmacology, Graduate Institute of Medicine, College of Medicine, Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Su-Ling Hsieh
- Department of Pharmacy, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Ing-Jun Chen
- Department of Pharmacology, Graduate Institute of Medicine, College of Medicine, Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Bin-Nan Wu
- Department of Pharmacology, Graduate Institute of Medicine, College of Medicine, Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
31
|
Understanding the mechanisms of angiotensin II signaling involved in hypertension and its long-term sequelae: insights from Bartter's and Gitelman's syndromes, human models of endogenous angiotensin II signaling antagonism. J Hypertens 2016; 32:2109-19; discussion 2119. [PMID: 25202962 DOI: 10.1097/hjh.0000000000000321] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Angiotensin II (Ang II) plays a key role in hypertension, renal and cardiovascular pathophysiology via intracellular pathways that involve the activation of a multiplicity of signaling mechanisms. Although experimental and genetic animal models have been developed and used to explore Ang II signaling's role in hypertension, a complete understanding of the processes mediating Ang II signaling in hypertension in humans remains elusive. One impediment is that these animal models do not exhibit all the traits of human hypertension, making it impossible to extrapolate from them to humans. To overcome this issue, we have used patients with Bartter's and Gitelman's syndromes, a human model of endogenously blunted and blocked Ang II signaling that presents a constellation of clinical findings which manifest themselves as the opposite of hypertension. This article reviews the aspects of the pathophysiology of human hypertension and its short and long term sequelae, and uses the results of our studies in Bartter's and Gitelman's syndromes along with those of others to gain better insight and understanding of the role of Ang II signaling in these processes.
Collapse
|
32
|
Ekman M, Albinsson S, Uvelius B, Swärd K. MicroRNAs in Bladder Outlet Obstruction: Relationship to Growth and Matrix Remodelling. Basic Clin Pharmacol Toxicol 2016; 119 Suppl 3:5-17. [DOI: 10.1111/bcpt.12534] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 11/12/2015] [Indexed: 12/22/2022]
Affiliation(s)
- Mari Ekman
- Department of Experimental Medical Science; Lund University; Lund Sweden
| | | | - Bengt Uvelius
- Department of Experimental Medical Science; Lund University; Lund Sweden
| | - Karl Swärd
- Department of Experimental Medical Science; Lund University; Lund Sweden
| |
Collapse
|
33
|
Aydinoglu F, Ergurhan Kiroglu O, Astarci E, Balli E, Ogulener N. Effects of ethanol on RhoA/Rho-kinase-mediated calcium sensitization in mouse lung parenchymal tissue. Eur J Pharmacol 2015; 764:318-327. [PMID: 26169563 DOI: 10.1016/j.ejphar.2015.07.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 06/29/2015] [Accepted: 07/08/2015] [Indexed: 11/16/2022]
Abstract
Calcium sensitization by the RhoA/Rho-kinase (ROCK) pathway contributes to the contraction in smooth muscle. Contractile stimuli can sensitize myosin to Ca(2+) by activating RhoA/Rho-kinase that inhibits myosin light chain phosphatase activity. The present study was aimed at investigating the possible involvement of RhoA/Rho-kinase pathway in contractile responses to agonist (phenylephrine) and depolarizing (KCl) of mouse lung parenchymal tissues. Also, we investigated the effect of ethanol on RhoA/Rho-kinase pathway. Phenylephrine (10(-8)-10(-4) M) and KCl (10-80 mM) induced sustained contractions in parenchymal strips. Ethanol significantly attenuated the contractions to phenylephrine and KCl. The Rho-kinase inhibitors fasudil (5×10(-5) M) and Y-27632 (5×10(-5) M) inhibited contractions to in both control and ethanol-treated parenchymal strips. In addition, the relaxations induced by fasudil (10(-4) M) and Y-27632 (5×10(-4) M) on parenchymal strips contracted by phenylephrine but not KCl was decreased in ethanol-treatment group. Also, RhoA, ROCK1 and ROCK2 expressions were detected in mouse lung parenchymal tissue. In ethanol-treated group, expression of RhoA and ROCK1 but not ROCK2 decreased compared to control. Furthermore, ethanol causes apoptotic changes in alveolar type I epithelial cells of parenchymal tissue. These results suggest that RhoA/Rho-kinase signaling pathway plays an important role in phenylephrine- and KCl-induced Ca(2)(+) sensitization in mouse lung parenchymal tissue. Also, ethanol may be decrease phenylephrine- and KCl-induced contraction due to lowering the RhoA/Rho-kinase-mediated Ca(2+)-sensitizing by inhibiting RhoA/Rho-kinase pathway in parenchymal tissue. These results may be lead to important insights into the mechanisms of lung diseases due to alcohol consumption.
Collapse
Affiliation(s)
- Fatma Aydinoglu
- Department of Pharmacology, Pharmacy Faculty, Cukurova University, Adana, Turkey
| | | | - Erhan Astarci
- Department of Plant and Animal Production, Mudurnu Süreyya Astarci Vocational School, Abant Izzet Baysal University, Bolu, Turkey
| | - Ebru Balli
- Department of Histology and Embryology, Medical Faculty, Mersin University, Mersin, Turkey
| | - Nuran Ogulener
- Department of Pharmacology, Medical Faculty, Cukurova University, Adana, Turkey.
| |
Collapse
|
34
|
Ravarotto V, Pagnin E, Maiolino G, Fragasso A, Carraro G, Rossi B, Calò LA. The blocking of angiotensin II type 1 receptor and RhoA/Rho kinase activity in hypertensive patients: Effect of olmesartan medoxomil and implication with cardiovascular-renal remodeling. J Renin Angiotensin Aldosterone Syst 2015; 16:1245-50. [DOI: 10.1177/1470320315594324] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 06/02/2015] [Indexed: 12/12/2022] Open
Affiliation(s)
- Verdiana Ravarotto
- Department of Medicine Nephrology, University of Padova-Azienda Ospedaliera Padova, Italy
- Hypertension Clinic, University of Padova-Azienda Ospedaliera Padova, Italy
| | - Elisa Pagnin
- Department of Medicine Nephrology, University of Padova-Azienda Ospedaliera Padova, Italy
| | - Giuseppe Maiolino
- Hypertension Clinic, University of Padova-Azienda Ospedaliera Padova, Italy
| | - Antonio Fragasso
- Department of Medicine Nephrology, University of Padova-Azienda Ospedaliera Padova, Italy
| | - Gianni Carraro
- Department of Medicine Nephrology, University of Padova-Azienda Ospedaliera Padova, Italy
| | - Barbara Rossi
- Department of Medicine Nephrology, University of Padova-Azienda Ospedaliera Padova, Italy
| | - Lorenzo A Calò
- Department of Medicine Nephrology, University of Padova-Azienda Ospedaliera Padova, Italy
| |
Collapse
|
35
|
Shibata K, Sakai H, Huang Q, Kamata H, Chiba Y, Misawa M, Ikebe R, Ikebe M. Rac1 regulates myosin II phosphorylation through regulation of myosin light chain phosphatase. J Cell Physiol 2015; 230:1352-64. [PMID: 25502873 DOI: 10.1002/jcp.24878] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 12/05/2014] [Indexed: 12/15/2022]
Abstract
Phosphorylation of regulatory light chain (MLC) activates myosin II, which enables it to promote contractile and motile activities of cells. We report here a novel signaling mechanism that activates MLC phosphorylation and smooth muscle contraction. Contractile agonists activated Rac1, and Rac1 inhibition diminished agonist-induced MLC phosphorylation, thus inhibiting smooth muscle contraction. Rac1 inhibits the activity of MLC phosphatase (MLCP) but not that of MLC kinase, through a phosphatase that targets MYPT1 (a regulatory subunit of MLCP) and CPI-17 (a MLCP specific inhibitor) rather than through the RhoA-Rho dependent kinase (ROCK) pathway. Rac1 inhibition decreased the activity of protein kinase C (PKC), which also contributes to the change in CPI-17 phosphorylation. We propose that activation of Rac1 increases the activity of PKC, which increases the phosphorylation of CPI-17 and MYPT1 by inhibiting the phosphatase that targets these proteins, thereby decreasing the activity of MLCP and increasing phosphorylation of MLC. Our results suggest that Rac1 coordinates with RhoA to increase MLC phosphorylation by inactivation of CPI-17/MYPT1 phosphatase, which decreases MLCP activity thus promoting MLC phosphorylation and cell contraction.
Collapse
Affiliation(s)
- Keita Shibata
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Albinsson S, Bhattachariya A, Hellstrand P. Stretch-dependent smooth muscle differentiation in the portal vein-role of actin polymerization, calcium signaling, and microRNAs. Microcirculation 2015; 21:230-8. [PMID: 24238368 DOI: 10.1111/micc.12106] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 11/09/2013] [Indexed: 11/28/2022]
Abstract
The mechanical forces acting on SMC in the vascular wall are known to regulate processes such as vascular remodeling and contractile differentiation. However, investigations to elucidate the underlying mechanisms of mechanotransduction in smooth muscle have been hampered by technical limitations associated with mechanical studies on pressurized small arteries, due primarily to the small amount of available tissue. The murine portal vein is a relatively large vessel showing myogenic tone that in many respects recapitulates the properties of small resistance vessels. Studies on stretched portal veins to elucidate mechanisms of mechanotransduction in the vascular wall have shown that stretch-sensitive regulation of contractile differentiation is mediated via Rho-activation and actin polymerization, while stretch-induced growth is regulated by the MAPK pathway. In this review, we have summarized findings on mechanotransduction in the portal vein with focus on stretch-induced contractile differentiation and the role of calcium, actin polymerization and miRNAs in this response.
Collapse
|
37
|
Wang B, Ding YM, Wang CT, Wang WX. Role of ROCK expression in gallbladder smooth muscle contraction. Mol Med Rep 2015; 12:2907-11. [PMID: 25955570 DOI: 10.3892/mmr.2015.3726] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Accepted: 04/10/2015] [Indexed: 11/06/2022] Open
Abstract
Cholelithiasis is a common medical condition whose incidence rate is increasing yearly, while its pathogenesis has yet to be elucidated. The present study assessed the expression of Rho-kinase (ROCK) in gallbladder smooth muscles and its effect on the contractile function of gallbladder smooth muscles during gallstone formation. Thirty male guinea pigs were randomly divided into three groups: The control group, the gallstone model group and the fasudil interference group. The fasting volume (FV) and bile capacity of the gallbladder (FB) as well as the total cholesterol (TC) and triglyceride (TG) contents of the gallbladder bile were determined. In addition, the gallbladder was dissected to identify whether any gallstones had formed. Part of the gallbladder tissue specimens were used for immunohistochemical analysis of ROCK expression in gallbladder smooth muscles. The results showed that four guinea pigs in the model group and eight in the fasudil group displayed gallstone formation, while there was no gallstone formation in the control group. The FV and FB were significantly increased in the model and fasudil groups. Similarly, the TC and TG contents of gallbladder bile were increased in these groups. The positive expression rate of ROCK in gallbladder smooth muscles in the model and fasudil groups was significantly reduced compared with that in the control group (P<0.05). The results of the present study indicated that the reduction of ROCK expression in guinea pig gallbladder smooth muscles weakened gallbladder contraction and thereby promoted gallstone formation.
Collapse
Affiliation(s)
- Bin Wang
- Department of Hepatobiliary and Laparoscopic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - You-Ming Ding
- Department of Hepatobiliary and Laparoscopic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Chun-Tao Wang
- Department of Hepatobiliary and Laparoscopic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Wei-Xing Wang
- Department of Hepatobiliary and Laparoscopic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
38
|
Increased activation of the Rho-A/Rho-kinase pathway in the renal vascular system is responsible for the enhanced reactivity to exogenous vasopressin in endotoxemic rats. Crit Care Med 2014; 42:e461-71. [PMID: 24690572 DOI: 10.1097/ccm.0000000000000313] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVE We evaluated the role of the renal vascular system and the Rho-A/Rho-kinase pathway in the maintenance of the pressor effects of vasopressin in endotoxemic rats. DESIGN In vitro and in vivo animal study. SETTING University research laboratory. SUBJECTS Male Wistar rats (200-300 g). INTERVENTION Rats received either saline or lipopolysaccharide (10 mg/kg, intraperitoneal) 6 or 24 hours before the experiments. The effects of vasopressin on isolated aortic rings, cardiac function, mean arterial pressure, and both the renal vascular perfusion pressure of perfused kidneys in vitro and renal blood flow in situ were evaluated. The role of Rho-kinase in the renal and systemic effects of vasopressin was investigated through administration of the selective inhibitor Y-27632 and Western blot analysis. MEASUREMENTS AND MAIN RESULTS The effect of vasopressin on mean arterial pressure was unaltered and that on renal vascular perfusion pressure enhanced in endotoxemic rats at both 6 and 24 hours after lipopolysaccharide, despite reduced contractile responses in aortic rings and the lack of effect on cardiac function. Vasopressin (3, 10, and 30 pmol/kg, IV) produced increased reduction in renal blood flow in endotoxemic rats. In perfused kidneys from lipopolysaccharide groups, administration of Y-27632 reverted the hyperreactivity to vasopressin. Treatment with Y-27632 partially inhibited the effects of vasopressin on mean arterial pressure and significantly reduced the effects of vasopressin on renal blood flow in control but not in endotoxemic rats. Although the protein levels of Rho-A and Rho-kinase I and II had not been impaired, the levels of phosphorylated myosin phosphatase-targeting subunit 1, the regulatory subunit of myosin phosphatase that is inhibited by Rho-kinase, were increased in both the renal cortex and the renal medulla of endotoxemic rats. CONCLUSION Our data suggest that activation of Rho-kinase potentiates the vascular effects of vasopressin in the kidneys, contributing to the maintenance of the hypertensive effects of this agent during septic shock.
Collapse
|
39
|
Protein kinase C in enhanced vascular tone in diabetes mellitus. Int J Cardiol 2014; 174:230-42. [DOI: 10.1016/j.ijcard.2014.04.117] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 04/03/2014] [Accepted: 04/09/2014] [Indexed: 12/24/2022]
|
40
|
Molecular and cellular basis of the regulation of lymphatic contractility and lymphatic absorption. Int J Biochem Cell Biol 2014; 53:134-40. [PMID: 24836907 DOI: 10.1016/j.biocel.2014.05.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 04/22/2014] [Accepted: 05/05/2014] [Indexed: 11/23/2022]
Abstract
Lymphatic absorption is a highly regulated process driven by both an extrinsic mechanism (external force) and an intrinsic mechanism (lymphatic vessel contractility). The lymphatic muscle is a specialized smooth muscle with unique mechanical properties. To understand the molecular mechanism and relative contribution of smooth muscle contraction in lymphatic absorption, we analyzed mice with a smooth muscle-specific deletion of Mylk, a critical gene for smooth muscle contraction. Interestingly, the knockout mice were significantly resistant to anesthesia reagents. Upon injection in the feet with FITC-dextran, the mutant mice displayed a 2-fold delay of the absorption peak in the peripheral circulation. Examining the ear lymphatic vessels of the mutant mice revealed a reduction in the amount of fluid in the lumens of the lymphangions, suggesting an impairment of lymph formation. The Mylk-deficient lymphatic muscle exhibited a significant reduction of peristalsis and of myosin light chain phosphorylation in response to depolarization. We thus concluded that MLCK and myosin light chain phosphorylation are required for lymphatic vessel contraction. Lymphatic contractility is not an exclusive requirement for lymphatic absorption, and external force appears to be necessary for absorption.
Collapse
|
41
|
Swärd K, Albinsson S, Rippe C. Arterial dysfunction but maintained systemic blood pressure in cavin-1-deficient mice. PLoS One 2014; 9:e92428. [PMID: 24658465 PMCID: PMC3962402 DOI: 10.1371/journal.pone.0092428] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 02/21/2014] [Indexed: 12/26/2022] Open
Abstract
Caveolae are omega-shaped plasma membrane micro-domains that are abundant in cells of the vascular system. Formation of caveolae depends on caveolin-1 and cavin-1 and lack of either protein leads to loss of caveolae. Mice with caveolin-1 deficiency have dysfunctional blood vessels, but whether absence of cavin-1 similarly leads to vascular dysfunction is not known. Here we addressed this hypothesis using small mesenteric arteries from cavin-1-deficient mice. Cavin-1-reporter staining was intense in mesenteric arteries, brain arterioles and elsewhere in the vascular system, with positive staining of both endothelial and smooth muscle cells. Arterial expression of cavin-1, -2 and -3 was reduced in knockout (KO) arteries as was expression of caveolin-1, -2 and -3. Caveolae were absent in the endothelial and smooth muscle layers of small mesenteric arteries as determined by electron microscopy. Arginase, a negative regulator of nitric oxide production, was elevated in cavin-1 deficient arteries as was contraction in response to the α1-adrenergic agonist cirazoline. Detailed assessment of vascular dimensions revealed increased media thickness and reduced distensibility, arguing that enhanced contraction was due to increased muscle mass. Contrasting with increased α1-adrenergic contraction, myogenic tone was essentially absent and this appeared to be due in part to increased nitric oxide production. Vasomotion was less frequent in the knock-out vessels. In keeping with the opposing influences on arterial resistance of increased agonist-induced contractility and reduced myogenic tone, arterial blood pressure was unchanged in vivo. We conclude that deficiency of cavin-1 affects the function of small arteries, but that opposing influences on arterial resistance balance each other such that systemic blood pressure in unstressed mice is well maintained.
Collapse
Affiliation(s)
- Karl Swärd
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | - Catarina Rippe
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
42
|
Mahavadi S, Sriwai W, Huang J, Grider JR, Murthy KS. Inhibitory signaling by CB1 receptors in smooth muscle mediated by GRK5/β-arrestin activation of ERK1/2 and Src kinase. Am J Physiol Gastrointest Liver Physiol 2014; 306:G535-45. [PMID: 24407588 PMCID: PMC3949025 DOI: 10.1152/ajpgi.00397.2013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
We examined whether CB1 receptors in smooth muscle conform to the signaling pattern observed with other Gi-coupled receptors that stimulate contraction via two Gβγ-dependent pathways (PLC-β3 and phosphatidylinositol 3-kinase/integrin-linked kinase). Here we show that the anticipated Gβγ-dependent signaling was abrogated. Except for inhibition of adenylyl cyclase via Gαi, signaling resulted from Gβγ-independent phosphorylation of CB1 receptors by GRK5, recruitment of β-arrestin1/2, and activation of ERK1/2 and Src kinase. Neither uncoupling of CB1 receptors from Gi by pertussis toxin (PTx) or Gi minigene nor expression of a Gβγ-scavenging peptide had any effect on ERK1/2 activity. The latter was abolished in muscle cells expressing β-arrestin1/2 siRNA. CB1 receptor internalization and both ERK1/2 and Src kinase activities were abolished in cells expressing kinase-deficient GRK5(K215R). Activation of ERK1/2 and Src kinase endowed CB1 receptors with the ability to inhibit concurrent contractile activity. We identified a consensus sequence (102KSPSKLSP109) for phosphorylation of RGS4 by ERK1/2 and showed that expression of a RGS4 mutant lacking Ser103/Ser108 blocked the ability of anandamide to inhibit acetylcholine-mediated phosphoinositide hydrolysis or enhance Gαq:RGS4 association and inactivation of Gαq. Activation of Src kinase by anandamide enhanced both myosin phosphatase RhoA-interacting protein (M-RIP):RhoA and M-RIP:MYPT1 association and inhibited Rho kinase activity, leading to increase of myosin light chain (MLC) phosphatase activity and inhibition of sustained muscle contraction. Thus, unlike other Gi-coupled receptors in smooth muscle, CB1 receptors did not engage Gβγ but signaled via GRK5/β-arrestin activation of ERK1/2 and Src kinase: ERK1/2 accelerated inactivation of Gαq by RGS4, and Src kinase enhanced MLC phosphatase activity, leading to inhibition of ACh-stimulated contraction.
Collapse
Affiliation(s)
- Sunila Mahavadi
- Department of Physiology and Biophysics, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Wimolpak Sriwai
- Department of Physiology and Biophysics, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Jiean Huang
- Department of Physiology and Biophysics, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - John R. Grider
- Department of Physiology and Biophysics, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Karnam S. Murthy
- Department of Physiology and Biophysics, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
43
|
Mita M, Tanaka H, Yanagihara H, Nakagawa JI, Hishinuma S, Sutherland C, Walsh MP, Shoji M. Membrane depolarization-induced RhoA/Rho-associated kinase activation and sustained contraction of rat caudal arterial smooth muscle involves genistein-sensitive tyrosine phosphorylation. J Smooth Muscle Res 2013; 49:26-45. [PMID: 24133693 PMCID: PMC5137315 DOI: 10.1540/jsmr.49.26] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Rho-associated kinase (ROK) activation plays an important role in K+-induced
contraction of rat caudal arterial smooth muscle (Mita et al., Biochem J. 2002; 364:
431–40). The present study investigated a potential role for tyrosine kinase activity in
K+-induced RhoA activation and contraction. The non-selective tyrosine kinase
inhibitor genistein, but not the src family tyrosine kinase inhibitor PP2, inhibited
K+-induced sustained contraction (IC50 = 11.3 ± 2.4 µM). Genistein
(10 µM) inhibited the K+-induced increase in myosin light chain
(LC20) phosphorylation without affecting the Ca2+ transient. The
tyrosine phosphatase inhibitor vanadate induced contraction that was reversed by genistein
(IC50 = 6.5 ± 2.3 µM) and the ROK inhibitor Y-27632 (IC50 = 0.27 ±
0.04 µM). Vanadate also increased LC20 phosphorylation in a genistein- and
Y-27632-dependent manner. K+ stimulation induced translocation of RhoA to the
membrane, which was inhibited by genistein. Phosphorylation of MYPT1 (myosin-targeting
subunit of myosin light chain phosphatase) was significantly increased at Thr855 and
Thr697 by K+ stimulation in a genistein- and Y-27632-sensitive manner. Finally,
K+ stimulation induced genistein-sensitive tyrosine phosphorylation of
proteins of ∼55, 70 and 113 kDa. We conclude that a genistein-sensitive tyrosine kinase,
activated by the membrane depolarization-induced increase in
[Ca2+]i, is involved in the RhoA/ROK activation and sustained
contraction induced by K+. Ca2+ sensitization, myosin light chain
phosphatase, RhoA, Rho-associated kinase, tyrosine kinase
Collapse
Affiliation(s)
- Mitsuo Mita
- Department of Pharmacodynamics, Meiji Pharmaceutical
University, Japan
| | - Hitoshi Tanaka
- Department of Pharmacodynamics, Meiji Pharmaceutical
University, Japan
| | - Hayato Yanagihara
- Department of Pharmacodynamics, Meiji Pharmaceutical
University, Japan
| | - Jun-ichi Nakagawa
- Department of Pharmacodynamics, Meiji Pharmaceutical
University, Japan
| | - Shigeru Hishinuma
- Department of Pharmacodynamics, Meiji Pharmaceutical
University, Japan
| | - Cindy Sutherland
- Smooth Muscle Research Group, Department of Biochemistry and
Molecular Biology, University of Calgary, Canada
| | - Michael P. Walsh
- Smooth Muscle Research Group, Department of Biochemistry and
Molecular Biology, University of Calgary, Canada
| | - Masaru Shoji
- Department of Pharmacodynamics, Meiji Pharmaceutical
University, Japan
| |
Collapse
|
44
|
Kendig DM, Matsumoto AK, Moreland RS. Sphingosine-1-phosphate induced contraction of bladder smooth muscle. Eur J Pharmacol 2013; 720:355-62. [PMID: 24120660 DOI: 10.1016/j.ejphar.2013.10.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 10/02/2013] [Accepted: 10/03/2013] [Indexed: 11/29/2022]
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid that contracts most smooth muscles. Although S1P has been shown to contract bladder smooth muscle, the mechanism(s) by which S1P initiates contraction has not been extensively investigated. The goal of this study was to determine if S1P-induced force generation and myosin light chain (MLC) phosphorylation are dependent on calcium sensitization pathways mediated by protein kinase C (PKC) and Rho kinase (ROCK) and which S1P receptor is important in this response. Bladder smooth muscle strips from rabbit and rat were mounted for isometric force recording and contracted in response to carbachol or S1P in the presence and absence of an inhibitor of PKC (3 µM Bisindolylmaleimide-1) or ROCK (1 µM H-1172). 10 µM S1P produced approximately 40% of the force generated in response to 110 mM KCl in rabbit bladder smooth muscle. S1P, up to 100 µM, did not produce a response in rat bladder smooth muscle, any response evoked was due to solvent (NaOH). S1P-dependent force development was associated with a concomitant increase in Ser(19), but not dual Thr(18)/Ser(19) MLC phosphorylation. Inhibition of PKC decreased force development, whereas inhibition of ROCK abolished S1P-induced force. An inhibitor of the S1P2 receptor, JTE-013, relaxed a S1P-induced contraction; whereas, an agonist with low affinity to the S1P2 receptor, dihydro-S1P, did not elicit a contraction. Our results suggest that S1P contracts rabbit, but not rat, bladder smooth muscle via the S1P2 receptor and is dependent on MLC phosphorylation and myofilament calcium sensitization primarily in response to ROCK activation.
Collapse
Affiliation(s)
- Derek M Kendig
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N 15th Street, MS #488, Philadelphia, PA 19102, USA
| | | | | |
Collapse
|
45
|
Zhang Y, Hermanson ME, Eddinger TJ. Tonic and phasic smooth muscle contraction is not regulated by the PKCα - CPI-17 pathway in swine stomach antrum and fundus. PLoS One 2013; 8:e74608. [PMID: 24058600 PMCID: PMC3776813 DOI: 10.1371/journal.pone.0074608] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Accepted: 08/04/2013] [Indexed: 01/31/2023] Open
Abstract
Regulation of myosin light chain phosphatase (MLCP) via protein kinase C (PKC) and the 17 kDa PKC-potentiated inhibitor of myosin light chain phosphatase (CPI-17) has been reported as a Ca2+ sensitization signaling pathway in smooth muscle (SM), and thus may be involved in tonic vs. phasic contractions. This study examined the protein expression and spatial-temporal distribution of PKCα and CPI-17 in intact SM tissues. KCl or carbachol (CCh) stimulation of tonic stomach fundus SM generates a sustained contraction while the phasic stomach antrum generates a transient contraction. In addition, the tonic fundus generates greater relative force than phasic antrum with 1 µM phorbol 12, 13-dibutyrate (PDBu) stimulation which is reported to activate the PKCα – CPI-17 pathway. Western blot analyses demonstrated that this contractile difference was not caused by a difference in the protein expression of PKCα or CPI-17 between these two tissues. Immunohistochemical results show that the distribution of PKCα in the longitudinal and circular layers of the fundus and antrum do not differ, being predominantly localized near the SM cell plasma membrane. Stimulation of either tissue with 1 µM PDBu or 1 µM CCh does not alter this peripheral PKCα distribution. There are no differences between these two tissues for the CPI-17 distribution, but unlike the PKCα distribution, CPI-17 appears to be diffusely distributed throughout the cytoplasm under relaxed tissue conditions but shifts to a primarily peripheral distribution at the plasma membrane with stimulation of the tissues with 1 µM PDBu or 1 µM CCh. Results from double labeling show that neither PKCα nor CPI-17 co-localize at the adherens junction (vinculin/talin) at the membrane but they do co-localize with each other and with caveoli (caveolin) at the membrane. This lack of difference suggests that the PKCα - CPI-17 pathway is not responsible for the tonic vs. phasic contractions observed in stomach fundus and antrum.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin, United States of America
| | - Meghan E. Hermanson
- Department of Biology, Bradley University, Peoria, Illinois, United States of America
| | - Thomas J. Eddinger
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
46
|
Wang J, Liu X, Zhong Y. Rho/Rho-associated kinase pathway in glaucoma (Review). Int J Oncol 2013; 43:1357-67. [PMID: 24042317 DOI: 10.3892/ijo.2013.2100] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 08/21/2013] [Indexed: 11/06/2022] Open
Abstract
The Rho/ROCK pathway plays important roles in the modulation of the cytoskeletal integrity of cells, the synthesis of extracellular matrix components in the aqueous humor outflow tissue and the permeability of Schlemm's canal endothelial cells. The activation of the Rho/ROCK pathway results in trabecular meshwork (TM) contraction, and the inhibition of this pathway would provoke relaxation of TM with subsequent increase in outflow facility and, thereby, decrease intraocular pressure (IOP). ROCK inhibitors also serve as potent anti‑scarring agents via inhibition of transdifferentiation of tenon fibroblasts into myofibroblasts. Furthermore, the RhoA/ROCK pathway is involved in optic nerve neuroprotection. Inactivation of Rho/ROCK signaling increase ocular blood flow, improve retinal ganglion cell (RGC) survival and promote RGC axon regeneration. Considering the IOP modulation, potent bleb anti-scarring effect and neuroprotective properties of ROCK inhibitors, the Rho/ROCK pathway is an attractive target for anti-glaucoma therapy, and it may be used for human therapy in the near future.
Collapse
Affiliation(s)
- Jing Wang
- Department of Ophthalmology, Ruijin Hospital Affiliated Medical School, Shanghai Jiaotong University, Shanghai 200025, P.R. China
| | | | | |
Collapse
|
47
|
Liu R, Zhang Z, Liu H, Hou P, Lang J, Wang S, Yan H, Li P, Huang Z, Wu H, Rong M, Huang J, Wang H, Lv L, Qiu M, Ding J, Lai R. Human β-defensin 2 is a novel opener of Ca2+-activated potassium channels and induces vasodilation and hypotension in monkeys. Hypertension 2013; 62:415-25. [PMID: 23734009 DOI: 10.1161/hypertensionaha.111.01076] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Human β-defensin 2 (HBD2) is a cysteine-rich cationic antimicrobial peptide known for its important role in innate immune system. Intensive studies have demonstrated its antimicrobial and chemotactic activities in vitro. In this study, ELISA analysis showed that HBD2 was significantly downregulated in sera of patients with hypertension. It relaxed vessel smooth muscle by acting on the major regulatory pathways, contributing to vessel smooth muscle contraction. Electrophysiology analysis indicated that HBD2 acted as an opener of large-conductance Ca(2+)-activated potassium (BKCa)-mSlo+hβ1 channels and increased BKCa currents. Mutation analysis revealed that HBD2 activated BKCa-mSlo+hβ1 channels via interacting with Leu41 and Gln43 of β1-loop. In vivo experiments suggested that HBD2 at 4 × to 6 × of physiological concentration exerted hypotensive effect in monkeys significantly, whereas the selective blocker of BKCa channels, Paxilline, inhibited the effect. HBD2 is the first peptide opener of BKCa-mSlo+hβ1 channels. It may be a novel regulator of blood pressure and provides a new therapeutic target for the treatment of hypertension. The HBD2 blockade of the BKCa channels may represent a new type of cross-talk between immune and cardiovascular systems.
Collapse
Affiliation(s)
- Rui Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Harraz OF, Welsh DG. T-Type Ca2+Channels in Cerebral Arteries: Approaches, Hypotheses, and Speculation. Microcirculation 2013; 20:299-306. [DOI: 10.1111/micc.12038] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Accepted: 01/07/2013] [Indexed: 01/08/2023]
Affiliation(s)
- Osama F. Harraz
- Department of Physiology & Pharmacology; Hotchkiss Brain and Libin Cardiovascular Research Institutes; University of Calgary; Calgary AB Canada
| | - Donald G. Welsh
- Department of Physiology & Pharmacology; Hotchkiss Brain and Libin Cardiovascular Research Institutes; University of Calgary; Calgary AB Canada
| |
Collapse
|
49
|
Shi XZ, Sarna SK. Cell culture retains contractile phenotype but epigenetically modulates cell-signaling proteins of excitation-contraction coupling in colon smooth muscle cells. Am J Physiol Gastrointest Liver Physiol 2013; 304:G337-45. [PMID: 23238936 PMCID: PMC3566616 DOI: 10.1152/ajpgi.00369.2012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Accepted: 12/11/2012] [Indexed: 01/31/2023]
Abstract
Smooth muscle cell cultures are used frequently to investigate the cellular mechanisms of contraction. We tested the hypothesis that cell culture alters the expression of select cell-signaling proteins of excitation-contraction coupling in colon smooth muscle cells without altering the contractile phenotype. We used muscularis externa (ME) tissues, freshly dispersed cells (FC), primary cell cultures (PC), and resuspensions of cell cultures (RC). Colon smooth muscle cells retained their phenotype in all states. We investigated expression of 10 cell-signaling proteins of excitation-contraction coupling in all four types of tissue. Expression of all these proteins did not differ between ME and FC (P > 0.05). However, expression of the α(1C)-subunit of Ca(v)1.2b, myosin light chain kinase, myosin phosphatase target subunit 1, and 17-kDa C kinase-potentiated protein phosphatase-1 inhibitor (CPI-17) decreased in PC and RC vs. ME and FC (all P < 0.05). Expression of Gα(i3), serine/threonine protein phosphatase-1 β-catalytic subunit, and Rho kinase 1 increased in PC and RC vs. ME and FC (all P < 0.05). Cell culture and resuspension downregulated expression of α-actin and calponin, but not myosin heavy chain. The net effect of these molecular changes was suppression of cell reactivity to ACh in RC vs. FC. Overexpression of CPI-17 in PC partially reversed the suppression of contractility in resuspended cells. Methylation-specific PCR showed increased methylation of the Cpi-17 gene promoter in PC vs. ME (P < 0.05). We concluded that smooth muscle cells retain their contractile phenotype in culture. However, reactivity to ACh declines because of altered expression of specific cell-signaling proteins involved in excitation-contraction coupling. DNA methylation of the Cpi-17 promoter may contribute to its gene suppression.
Collapse
Affiliation(s)
- Xuan-Zheng Shi
- Enteric Neuromuscular Disorders and Visceral Pain Center, Division of Gastroenterology, Department of Internal Medicine, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | | |
Collapse
|
50
|
Affiliation(s)
- Jonathan D Schiff
- James Buchanan Brady Foundation, Department of Urology, New York-Weill Cornell Medical Center, New York, New York 10021, USA
| | | |
Collapse
|