151
|
Inhibition of ROCK2 alleviates renal fibrosis and the metabolic disorders in the proximal tubular epithelial cells. Clin Sci (Lond) 2020; 134:1357-1376. [PMID: 32490513 DOI: 10.1042/cs20200030] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 06/01/2020] [Accepted: 06/02/2020] [Indexed: 12/12/2022]
Abstract
Non-specific inhibition of Rho-associated kinases (ROCKs) alleviated renal fibrosis in the unilateral ureteral obstruction (UUO) model, while genetic deletion of ROCK1 did not affect renal pathology in mice. Thus, whether ROCK2 plays a role in renal tubulointerstitial fibrosis needs to be clarified. In the present study, a selective inhibitor against ROCK2 or genetic approach was used to investigate the role of ROCK2 in renal tubulointerstitial fibrosis. In the fibrotic kidneys of chronic kidney diseases (CKDs) patients, we observed an enhanced expression of ROCK2 with a positive correlation with interstitial fibrosis. In mice, the ROCK2 protein level was time-dependently increased in the UUO model. By treating CKD animals with KD025 at the dosage of 50 mg/kg/day via intraperitoneal injection, the renal fibrosis shown by Masson's trichrome staining was significantly alleviated along with the reduced expression of fibrotic genes. In vitro, inhibiting ROCK2 by KD025 or ROCK2 knockdown/knockout significantly blunted the pro-fibrotic response in transforming growth factor-β1 (TGF-β1)-stimulated mouse renal proximal tubular epithelial cells (mPTCs). Moreover, impaired cellular metabolism was reported as a crucial pathogenic factor in CKD. By metabolomics analysis, we found that KD025 restored the metabolic disturbance, including the impaired glutathione metabolism in TGF-β1-stimulated tubular epithelial cells. Consistently, KD025 increased antioxidative stress enzymes and nuclear erythroid 2-related factor 2 (Nrf2) in fibrotic models. In addition, KD025 decreased the infiltration of macrophages and inflammatory response in fibrotic kidneys and blunted the activation of macrophages in vitro. In conclusion, inhibition of ROCK2 may serve as a potential novel therapy for renal tubulointerstitial fibrosis in CKD.
Collapse
|
152
|
Matoba K, Takeda Y, Nagai Y, Sekiguchi K, Yokota T, Utsunomiya K, Nishimura R. The Physiology, Pathology, and Therapeutic Interventions for ROCK Isoforms in Diabetic Kidney Disease. Front Pharmacol 2020; 11:585633. [PMID: 33101039 PMCID: PMC7545791 DOI: 10.3389/fphar.2020.585633] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/07/2020] [Indexed: 01/14/2023] Open
Abstract
Rho-associated coiled-coil-containing protein kinase (ROCK) is a serine/threonine kinase that was originally identified as RhoA interacting protein. A diverse array of cellular functions, including migration, proliferation, and phenotypic modulation, are orchestrated by ROCK through a mechanism involving cytoskeletal rearrangement. Mammalian cells express two ROCK isoforms: ROCK1 (Rho-kinase β/ROKβ) and ROCK2 (Rho-kinase α/ROKα). While both isoforms have structural similarities and are widely expressed across multiple tissues, investigations in gene knockout animals and cell-based studies have revealed distinct functions of ROCK1 and ROCK2. With respect to the kidney, inhibiting ROCK activity has proven effective for the preventing diabetic kidney disease (DKD) in both type 1 and type 2 diabetic rodent models. However, despite significant progress in the understanding of the renal ROCK biology over the past decade, the pathogenic roles of the ROCK isoforms is only beginning to be elucidated. Recent studies have demonstrated the involvement of renal ROCK1 in mitochondrial dynamics and cellular transdifferentiation, whereas ROCK2 activation leads to inflammation, fibrosis, and cell death in the diabetic kidney. This review provides a conceptual framework for dissecting the molecular underpinnings of ROCK-driven renal injury, focusing on the differences between ROCK1 and ROCK2.
Collapse
Affiliation(s)
- Keiichiro Matoba
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Yusuke Takeda
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Yosuke Nagai
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Kensuke Sekiguchi
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Tamotsu Yokota
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Kazunori Utsunomiya
- Center for Preventive Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Rimei Nishimura
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
153
|
Zhang Z, Nong L, Chen M, Gu X, Zhao W, Liu M, Cheng W. Baicalein suppresses vasculogenic mimicry through inhibiting RhoA/ROCK expression in lung cancer A549 cell line. Acta Biochim Biophys Sin (Shanghai) 2020; 52:1007-1015. [PMID: 32672788 DOI: 10.1093/abbs/gmaa075] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/06/2020] [Accepted: 03/09/2020] [Indexed: 12/19/2022] Open
Abstract
Vasculogenic mimicry (VM) refers to a new tubular network of the blood supply system with abundant extracellular matrix. VM is similar to capillaries but does not involve endothelial cells. As a traditional herbal medicine commonly used in China, baicalein possesses anti-inflammatory and lipoxygenase activities. However, the effects of baicalein on the process of VM formation in non-small cell lung cancer (NSCLC) and the underlying mechanisms have remained poorly understood. In this study, baicalein was found to inhibit the viability and motility of A549 cells and induced the breakage of the cytoskeletal actin filament network. In addition, baicalein significantly decreased the formation of VM and downregulated the expressions of VM-associated factors, such as VE-cadherin, EphA2, MMP14, MMP2, MMP9, PI3K and LAMC2, similar to the effects of ROCK inhibitors. Indeed, baicalein inhibited RhoA/ROCK expression in vitro and in vivo, suggesting the underlying mechanisms of reduced VM formation. Collectively, baicalein suppressed the formation of VM in NSCLC by targeting the RhoA/ROCK signaling pathway, indicating that baicalein might serve as an emerging drug for NSCLC.
Collapse
Affiliation(s)
- Zhe Zhang
- Department of Integrated Therapy, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China, and
| | - Li Nong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Menglei Chen
- Department of Integrated Therapy, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China, and
| | - Xiaoli Gu
- Department of Integrated Therapy, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China, and
| | - Weiwei Zhao
- Department of Integrated Therapy, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China, and
| | - Minghui Liu
- Department of Integrated Therapy, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China, and
| | - Wenwu Cheng
- Department of Integrated Therapy, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China, and
| |
Collapse
|
154
|
Moretti R, Caruso P. An Iatrogenic Model of Brain Small-Vessel Disease: Post-Radiation Encephalopathy. Int J Mol Sci 2020; 21:6506. [PMID: 32899565 PMCID: PMC7555594 DOI: 10.3390/ijms21186506] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/27/2020] [Accepted: 09/01/2020] [Indexed: 12/21/2022] Open
Abstract
We studied 114 primitive cerebral neoplasia, that were surgically treated, and underwent radiotherapy (RT), and compared their results to those obtained by 190 patients diagnosed with subcortical vascular dementia (sVAD). Patients with any form of primitive cerebral neoplasia underwent whole-brain radiotherapy. All the tumor patients had regional field partial brain RT, which encompassed each tumor, with an average margin of 2.6 cm from the initial target tumor volume. We observed in our patients who have been exposed to a higher dose of RT (30-65 Gy) a cognitive and behavior decline similar to that observed in sVAD, with the frontal dysexecutive syndrome, apathy, and gait alterations, but with a more rapid onset and with an overwhelming effect. Multiple mechanisms are likely to be involved in radiation-induced cognitive impairment. The active site of RT brain damage is the white matter areas, particularly the internal capsule, basal ganglia, caudate, hippocampus, and subventricular zone. In all cases, radiation damage inside the brain mainly focuses on the cortical-subcortical frontal loops, which integrate and process the flow of information from the cortical areas, where executive functions are "elaborated" and prepared, towards the thalamus, subthalamus, and cerebellum, where they are continuously refined and executed. The active mechanisms that RT drives are similar to those observed in cerebral small vessel disease (SVD), leading to sVAD. The RT's primary targets, outside the tumor mass, are the blood-brain barrier (BBB), the small vessels, and putative mechanisms that can be taken into account are oxidative stress and neuro-inflammation, strongly associated with the alteration of NMDA receptor subunit composition.
Collapse
Affiliation(s)
- Rita Moretti
- Department Medical, Surgical, Health Sciences, University of Trieste, Cattinara Hospital, Strada di Fiume, 447, 34149 Trieste, Italy;
- Neurological Clinic, Department of Internal Medicine and Neurology, University of Trieste, Cattinara Hospital, Strada di Fiume, 447, 34149 Trieste, Italy
| | - Paola Caruso
- Department Medical, Surgical, Health Sciences, University of Trieste, Cattinara Hospital, Strada di Fiume, 447, 34149 Trieste, Italy;
- Neurological Clinic, Department of Internal Medicine and Neurology, University of Trieste, Cattinara Hospital, Strada di Fiume, 447, 34149 Trieste, Italy
| |
Collapse
|
155
|
Mulindwa J, Noyes H, Ilboudo H, Pagani L, Nyangiri O, Kimuda MP, Ahouty B, Asina OF, Ofon E, Kamoto K, Kabore JW, Koffi M, Ngoyi DM, Simo G, Chisi J, Sidibe I, Enyaru J, Simuunza M, Alibu P, Jamonneau V, Camara M, Tait A, Hall N, Bucheton B, MacLeod A, Hertz-Fowler C, Matovu E, Matovu E, Sidibe I, Mumba D, Koffi M, Simo G, Chisi J, Alibu VP, Macleod A, Bucheton B, Hertzfowler C, Elliot A, Camara M, Bishop O, Mulindwa J, Nyangiri O, Kimuda MP, Ofon E, Ahouty B, Kabore J. High Levels of Genetic Diversity within Nilo-Saharan Populations: Implications for Human Adaptation. Am J Hum Genet 2020; 107:473-486. [PMID: 32781046 PMCID: PMC7477016 DOI: 10.1016/j.ajhg.2020.07.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 07/13/2020] [Indexed: 12/23/2022] Open
Abstract
Africa contains more human genetic variation than any other continent, but the majority of the population-scale analyses of the African peoples have focused on just two of the four major linguistic groups, the Niger-Congo and Afro-Asiatic, leaving the Nilo-Saharan and Khoisan populations under-represented. In order to assess genetic variation and signatures of selection within a Nilo-Saharan population and between the Nilo-Saharan and Niger-Congo and Afro-Asiatic, we sequenced 50 genomes from the Nilo-Saharan Lugbara population of North-West Uganda and 250 genomes from 6 previously unsequenced Niger-Congo populations. We compared these data to data from a further 16 Eurasian and African populations including the Gumuz, another putative Nilo-Saharan population from Ethiopia. Of the 21 million variants identified in the Nilo-Saharan population, 3.57 million (17%) were not represented in dbSNP and included predicted non-synonymous mutations with possible phenotypic effects. We found greater genetic differentiation between the Nilo-Saharan Lugbara and Gumuz populations than between any two Afro-Asiatic or Niger-Congo populations. F3 tests showed that Gumuz contributed a genetic component to most Niger-Congo B populations whereas Lugabara did not. We scanned the genomes of the Lugbara for evidence of selective sweeps. We found selective sweeps at four loci (SLC24A5, SNX13, TYRP1, and UVRAG) associated with skin pigmentation, three of which already have been reported to be under selection. These selective sweeps point toward adaptations to the intense UV radiation of the Sahel.
Collapse
|
156
|
Freitas-Andrade M, Raman-Nair J, Lacoste B. Structural and Functional Remodeling of the Brain Vasculature Following Stroke. Front Physiol 2020; 11:948. [PMID: 32848875 PMCID: PMC7433746 DOI: 10.3389/fphys.2020.00948] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 07/14/2020] [Indexed: 12/12/2022] Open
Abstract
Maintenance of cerebral blood vessel integrity and regulation of cerebral blood flow ensure proper brain function. The adult human brain represents only a small portion of the body mass, yet about a quarter of the cardiac output is dedicated to energy consumption by brain cells at rest. Due to a low capacity to store energy, brain health is heavily reliant on a steady supply of oxygen and nutrients from the bloodstream, and is thus particularly vulnerable to stroke. Stroke is a leading cause of disability and mortality worldwide. By transiently or permanently limiting tissue perfusion, stroke alters vascular integrity and function, compromising brain homeostasis and leading to widespread consequences from early-onset motor deficits to long-term cognitive decline. While numerous lines of investigation have been undertaken to develop new pharmacological therapies for stroke, only few advances have been made and most clinical trials have failed. Overall, our understanding of the acute and chronic vascular responses to stroke is insufficient, yet a better comprehension of cerebrovascular remodeling following stroke is an essential prerequisite for developing novel therapeutic options. In this review, we present a comprehensive update on post-stroke cerebrovascular remodeling, an important and growing field in neuroscience, by discussing cellular and molecular mechanisms involved, sex differences, limitations of preclinical research design and future directions.
Collapse
Affiliation(s)
| | - Joanna Raman-Nair
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Baptiste Lacoste
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
157
|
Rho-Kinase inhibitors ameliorate diclofenac-induced cardiotoxicity in chloroquine-treated adjuvant arthritic rats. Life Sci 2020; 254:117605. [DOI: 10.1016/j.lfs.2020.117605] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 03/24/2020] [Accepted: 03/27/2020] [Indexed: 12/15/2022]
|
158
|
Wang J, Du A, Wang H, Li Y. MiR-599 regulates LPS-mediated apoptosis and inflammatory responses through the JAK2/STAT3 signalling pathway via targeting ROCK1 in human umbilical vein endothelial cells. Clin Exp Pharmacol Physiol 2020; 47:1420-1428. [PMID: 32248560 DOI: 10.1111/1440-1681.13316] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 03/21/2020] [Accepted: 03/24/2020] [Indexed: 12/17/2022]
Abstract
MicroRNA plays an integral role in the development of atherosclerosis. Our study aimed to investigate the roles of miR-599 in lipopolysaccharide (LPS)-induced endothelial damage in human umbilical vein endothelial cells (HUVECs). HUVECs were transfected with a miR-599 mimic and negative control, and then exposed to LPS. The expression of miR-599 was detected by quantitative real time-polymerase chain reaction (RT-qPCR). Cell viability was analyzed by CCK-8 assay and trypan blue exclusion assay; the formation of DNA fragments was tested by Cell Death Detection ELISA Plus kit; the incidence of apoptosis was detected by flow cytometry; the expression of p53 and cleaved-caspase 3 (c-caspase 3) was evaluated by western blot. Moreover, the mRNA levels and concentrations of tumour necrosis factor (TNF)-α, interleukin (IL)-6, ICAM-1 and VCAM-1 were assayed by RT-qPCR and ELISA. The results showed that overexpression of miR-599 increased cell viability, reduced DNA fragments, the incidence of apoptosis, as well as the protein levels of p53 and c-caspase 3 in the presence of LPS. TNF-α, IL-6, ICAM-1 and VCAM-1 mRNA levels and concentrations were also decreased upon miR-599 upregulation. In addition, the dual luciferase reporter assay demonstrated that ROCK1 is a direct target of miR-599. MiR-599 overexpression inhibited ROCK1 expression. Induced expression of ROCK1 reversed the roles of miR-599 in apoptosis and inflammation. The gain function of miR-599 function inhibited activation of the JAK2/STAT3 signalling pathway, which was abrogated by overexpression of ROCK1. Taken together, our results indicate that miR-599 attenuates LPS-caused cell apoptosis and inflammatory responses through the JAK2/STAT3 signalling pathway via targeting ROCK1.
Collapse
Affiliation(s)
- Jia Wang
- Department of Cardiology, Nursing Department, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Aolin Du
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Hexilin Wang
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yang Li
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
159
|
Maekawa M, Higashiyama S. KCTD10 Biology: An Adaptor for the Ubiquitin E3 Complex Meets Multiple Substrates: Emerging Divergent Roles of the cullin-3/KCTD10 E3 Ubiquitin Ligase Complex in Various Cell Lines. Bioessays 2020; 42:e1900256. [PMID: 32484264 DOI: 10.1002/bies.201900256] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 04/08/2020] [Indexed: 12/18/2022]
Abstract
Protein ubiquitination constitutes a post-translational modification mediated by ubiquitin ligases whereby ubiquitinated substrates are degraded through the proteasomal or lysosomal pathways, or acquire novel molecular functions according to their "ubiquitin codes." Dysfunction of the ubiquitination process in cells causes various diseases such as cancers along with neurodegenerative, auto-immune/inflammatory, and metabolic diseases. KCTD10 functions as a substrate recognition receptor for cullin-3 (CUL3), a scaffold protein in RING-type ubiquitin ligase complexes. Recently, studies by ourselves and others have identified new substrates that are ubiquitinated by the CUL3/KCTD10 ubiquitin ligase complex. Moreover, the type of polyubiquitination (e.g., K27-, K48-, or K63-chain) of various substrates (e.g., RhoB, CEP97, EIF3D, and TRIF) mediated by KCTD10 underlies its divergent roles in endothelial barrier formation, primary cilium formation, plasma membrane dynamics, cell proliferation, and immune response. Here, the physiological functions of KCTD10 are summarized and potential mechanisms are proposed.
Collapse
Affiliation(s)
- Masashi Maekawa
- Division of Cell Growth and Tumor Regulation, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime, 791-0295, Japan.,Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime, 791-0295, Japan
| | - Shigeki Higashiyama
- Division of Cell Growth and Tumor Regulation, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime, 791-0295, Japan.,Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime, 791-0295, Japan
| |
Collapse
|
160
|
Yuan Y, Khan S, Stewart DJ, Courtman DW. Engineering blood outgrowth endothelial cells to optimize endothelial nitric oxide synthase and extracellular matrix production for coating of blood contacting surfaces. Acta Biomater 2020; 109:109-120. [PMID: 32302726 DOI: 10.1016/j.actbio.2020.04.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 04/06/2020] [Accepted: 04/08/2020] [Indexed: 12/26/2022]
Abstract
Coverage of blood contacting surfaces by a functional endothelial layer is likely required to induce and maintain homeostasis. Blood outgrowth endothelial cells (BOECs), cultured from human peripheral blood monocytes, are readily available and functional autologous endothelial source that may represent a reasonable alternative to vascular derived cells. Endothelial nitric oxide synthase (eNOS) produces NO, an important factor that regulates homeostasis at the blood-contacting surface. We found that BOECs express markedly lower levels of eNOS protein (34% ± 13%, Western blot) and mRNA (29% ± 17%, qRT-PCR), as well as exhibiting reduced activity (49% ± 18%, Nitrite analysis) when compared to human umbilical vein endothelial cells (HUVECs) and human aortic endothelial cells. HUVECs grown on fibronectin, type I collagen, or laminin -coated surfaces exhibited significant reduction of eNOS mRNA and protein expression. However, no decrease in eNOS levels was observed in BOECs. Interestingly BOECs expressed significantly higher Collagen (Col) I compared to HUVECs, and blocking Col I synthesis significantly enhanced eNOS expression in BOECs. Inhibition of β1 integrin, focal adhesion kinase (FAK), or actin polymerization increased eNOS in both BOECs and HUVECs suggesting involvement of a signaling pathway culminating in stabilization of the cytoskeleton. Finally, we demonstrated that a Rho-associated protein kinases (ROCK) inhibitor, as a disruptor of actin stabilization, enhanced both eNOS expression and bioactivity. Taken together, our findings demonstrate that cell-ECM interactions are fundamental to the regulation of eNOS in BOECs and suggest that disruption of key intracellular pathways (such as ROCK) may be necessary to enhance functional activity of an endothelialized surface. STATEMENT OF SIGNIFICANCE: Development of biocompatible blood-contacting biomaterial surfaces has not been possible to date, leading many investigators to believe that a complete autologous endothelial layer will be necessary. Blood outgrowth endothelial cells (BOECs), cultured from human peripheral blood monocytes, are readily available and functional autologous endothelial source. Endothelial nitric oxide synthase (eNOS) produces NO, an important factor that regulates homeostasis at the blood-contacting surface. In this study, we show that eNOS displays limited expression in cultured BOECs. We further demonstrate that a strong negative regulation of eNOS is mediated by collagen substrates and that treatment with ROCK inhibitor could enhance both eNOS expression and activity in BOECs and help to rapidly establish a functional autologous endothelial layer on cardiovascular biomaterials.
Collapse
Affiliation(s)
- Yifan Yuan
- Ottawa Hospital Research Institute, General Campus, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; Department of Anaesthesiology, Yale University, 10 Amistad Rd, New Haven, CT 06519, United States
| | - Saad Khan
- Ottawa Hospital Research Institute, General Campus, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada
| | - Duncan J Stewart
- Ottawa Hospital Research Institute, General Campus, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; Department of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - David W Courtman
- Ottawa Hospital Research Institute, General Campus, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada; Department of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada.
| |
Collapse
|
161
|
Harnessing the Power of Eph/ephrin Biosemiotics for Theranostic Applications. Pharmaceuticals (Basel) 2020; 13:ph13060112. [PMID: 32492868 PMCID: PMC7345574 DOI: 10.3390/ph13060112] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 05/26/2020] [Accepted: 05/27/2020] [Indexed: 02/07/2023] Open
Abstract
Comprehensive basic biological knowledge of the Eph/ephrin system in the physiologic setting is needed to facilitate an understanding of its role and the effects of pathological processes on its activity, thereby paving the way for development of prospective therapeutic targets. To this end, this review briefly addresses what is currently known and being investigated in order to highlight the gaps and possible avenues for further investigation to capitalize on their diverse potential.
Collapse
|
162
|
Jin Y, Blikslager AT. The Regulation of Intestinal Mucosal Barrier by Myosin Light Chain Kinase/Rho Kinases. Int J Mol Sci 2020; 21:ijms21103550. [PMID: 32443411 PMCID: PMC7278945 DOI: 10.3390/ijms21103550] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/15/2020] [Accepted: 05/16/2020] [Indexed: 12/19/2022] Open
Abstract
The intestinal epithelial apical junctional complex, which includes tight and adherens junctions, contributes to the intestinal barrier function via their role in regulating paracellular permeability. Myosin light chain II (MLC-2), has been shown to be a critical regulatory protein in altering paracellular permeability during gastrointestinal disorders. Previous studies have demonstrated that phosphorylation of MLC-2 is a biochemical marker for perijunctional actomyosin ring contraction, which increases paracellular permeability by regulating the apical junctional complex. The phosphorylation of MLC-2 is dominantly regulated by myosin light chain kinase- (MLCK-) and Rho-associated coiled-coil containing protein kinase- (ROCK-) mediated pathways. In this review, we aim to summarize the current state of knowledge regarding the role of MLCK- and ROCK-mediated pathways in the regulation of the intestinal barrier during normal homeostasis and digestive diseases. Additionally, we will also suggest potential therapeutic targeting of MLCK- and ROCK-associated pathways in gastrointestinal disorders that compromise the intestinal barrier.
Collapse
Affiliation(s)
- Younggeon Jin
- Department of Animal and Avian Sciences, College of Agriculture and Natural Resources, University of Maryland, College Park, MD 20742, USA;
| | - Anthony T. Blikslager
- Department of Clinical Sciences, Comparative Medicine Institute, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
- Correspondence:
| |
Collapse
|
163
|
Seccia TM, Rigato M, Ravarotto V, Calò LA. ROCK (RhoA/Rho Kinase) in Cardiovascular-Renal Pathophysiology: A Review of New Advancements. J Clin Med 2020; 9:jcm9051328. [PMID: 32370294 PMCID: PMC7290501 DOI: 10.3390/jcm9051328] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 04/28/2020] [Accepted: 04/29/2020] [Indexed: 12/11/2022] Open
Abstract
Rho-associated, coiled-coil containing kinases (ROCK) were originally identified as effectors of the RhoA small GTPase and found to belong to the AGC family of serine/threonine kinases. They were shown to be downstream effectors of RhoA and RhoC activation. They signal via phosphorylation of proteins such as MYPT-1, thereby regulating many key cellular functions including proliferation, motility and viability and the RhoA/ROCK signaling has been shown to be deeply involved in arterial hypertension, cardiovascular–renal remodeling, hypertensive nephropathy and posttransplant hypertension. Given the deep involvement of ROCK in cardiovascular–renal pathophysiology and the interaction of ROCK signaling with other signaling pathways, the reports of trials on the clinical beneficial effects of ROCK’s pharmacologic targeting are growing. In this current review, we provide a brief survey of the current understanding of ROCK-signaling pathways, also integrating with the more novel data that overall support a relevant role of ROCK for the cardiovascular–renal physiology and pathophysiology.
Collapse
Affiliation(s)
- Teresa M. Seccia
- Department of Medicine, Hypertension Clinic, University of Padova, 35128 Padova, Italy;
| | - Matteo Rigato
- Department of Medicine, Nephrology, Dialysis and Transplantation Unit, University of Padova, 35128 Padova, Italy; (M.R.); (V.R.)
| | - Verdiana Ravarotto
- Department of Medicine, Nephrology, Dialysis and Transplantation Unit, University of Padova, 35128 Padova, Italy; (M.R.); (V.R.)
| | - Lorenzo A. Calò
- Department of Medicine, Nephrology, Dialysis and Transplantation Unit, University of Padova, 35128 Padova, Italy; (M.R.); (V.R.)
- Correspondence: ; Tel.: +39-049-8213071; Fax: +39-049-8217921
| |
Collapse
|
164
|
Determination of KD025 (SLx-2119), a Selective ROCK2 Inhibitor, in Rat Plasma by High-Performance Liquid Chromatography-Tandem Mass Spectrometry and its Pharmacokinetic Application. Molecules 2020; 25:molecules25061369. [PMID: 32192179 PMCID: PMC7144358 DOI: 10.3390/molecules25061369] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/13/2020] [Accepted: 03/13/2020] [Indexed: 12/22/2022] Open
Abstract
KD025 (SLx-2119), the first specific Rho-associated protein kinase 2 (ROCK2) inhibitor, is a potential new drug candidate currently undergoing several phase 2 clinical trials for psoriasis, idiopathic pulmonary fibrosis, chronic graft-versus-host disease, and systemic sclerosis. In this study, a bio-analytical method was developed and fully validated for the quantification of KD025 in rat plasma and for application in pharmacokinetic studies. KD025 and GSK429286A (the internal standard) in rat plasma samples were analyzed by high-performance liquid chromatography-tandem mass spectrometry with m/z transition values of 453.10 → 366.10 and 433.00 → 178.00, respectively. The method was fully validated according to the United State Food and Drug Administration guidelines in terms of selectivity, linearity, accuracy, precision, sensitivity, matrix effects, extraction recovery, and stability. The method enabled the quantification of KD025 levels in rat plasma following oral administration of 5 mg/kg KD025 and intravenous administration of 2 mg/kg KD025 to rats, respectively. Our findings suggest that the developed method is practical and reliable for pharmacokinetic studies of KD025 in preclinical animals.
Collapse
|
165
|
Haploinsufficient Rock1+/- and Rock2+/- Mice Are Not Protected from Cardiac Inflammation and Postinflammatory Fibrosis in Experimental Autoimmune Myocarditis. Cells 2020; 9:cells9030700. [PMID: 32178482 PMCID: PMC7140701 DOI: 10.3390/cells9030700] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/02/2020] [Accepted: 03/11/2020] [Indexed: 12/18/2022] Open
Abstract
Progressive cardiac fibrosis is a common cause of heart failure. Rho-associated, coiled-coil-containing protein kinases (ROCKs) have been shown to enhance fibrotic processes in the heart and in other organs. In this study, using wild-type, Rock1+/− and Rock2+/− haploinsufficient mice and mouse model of experimental autoimmune myocarditis (EAM) we addressed the role of ROCK1 and ROCK2 in development of myocarditis and postinflammatory fibrosis. We found that myocarditis severity was comparable in wild-type, Rock1+/− and Rock2+/− mice at day 21 of EAM. During the acute stage of the disease, hearts of Rock1+/− mice showed unaffected numbers of CD11b+CD36+ macrophages, CD11b+CD36–Ly6GhiLy6chi neutrophils, CD11b+CD36–Ly6G–Ly6chi inflammatory monocytes, CD11b+CD36–Ly6G–Ly6c– monocytes, CD11b+SiglecF+ eosinophils, CD11b+CD11c+ inflammatory dendritic cells and type I collagen-producing fibroblasts. Isolated Rock1+/− cardiac fibroblasts treated with transforming growth factor-beta (TGF-β) showed attenuated Smad2 and extracellular signal-regulated kinase (Erk) phosphorylations that were associated with impaired upregulation of smooth muscle actin alpha (αSMA) protein. In contrast to cardiac fibroblasts, expanded Rock1+/− heart inflammatory myeloid cells showed unaffected Smad2 activation but enhanced Erk phosphorylation following TGF-β treatment. Rock1+/− inflammatory cells responded to TGF-β by a reduced transcriptional profibrotic response and failed to upregulate αSMA and fibronectin at the protein levels. Unexpectedly, in the EAM model wild-type, Rock1+/− and Rock2+/− mice developed a similar extent of cardiac fibrosis at day 40. In addition, hearts of the wild-type and Rock1+/− mice showed comparable levels of cardiac vimentin, periostin and αSMA. In conclusion, despite the fact that ROCK1 regulates TGF-β-dependent profibrotic response, neither ROCK1 nor ROCK2 is critically involved in the development of postinflammatory fibrosis in the EAM model.
Collapse
|
166
|
Bryson TD, Pandrangi TS, Khan SZ, Xu J, Pavlov TS, Ortiz PA, Peterson E, Harding P. The deleterious role of the prostaglandin E 2 EP 3 receptor in angiotensin II hypertension. Am J Physiol Heart Circ Physiol 2020; 318:H867-H882. [PMID: 32142358 DOI: 10.1152/ajpheart.00538.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Angiotensin II (ANG II) plays a key role in regulating blood pressure and inflammation. Prostaglandin E2 (PGE2) signals through four different G protein-coupled receptors, eliciting a variety of effects. We reported that activation of the EP3 receptor reduces cardiac contractility. More recently, we have shown that overexpression of the EP4 receptor is protective in a mouse myocardial infarction model. We hypothesize in this study that the relative abundance of EP3 and EP4 receptors is a major determinant of end-organ damage in the diseased heart. Thus EP3 is detrimental to cardiac function and promotes inflammation, whereas antagonism of the EP3 receptor is protective in an ANG II hypertension (HTN) model. To test our hypothesis, male 10- to 12-wk-old C57BL/6 mice were anesthetized with isoflurane and osmotic minipumps containing ANG II were implanted subcutaneously for 2 wk. We found that antagonism of the EP3 receptor using L798,106 significantly attenuated the increase in blood pressure with ANG II infusion. Moreover, antagonism of the EP3 receptor prevented a decline in cardiac function after ANG II treatment. We also found that 10- to 12-wk-old EP3-transgenic mice, which overexpress EP3 in the cardiomyocytes, have worsened cardiac function. In conclusion, activation or overexpression of EP3 exacerbates end-organ damage in ANG II HTN. In contrast, antagonism of the EP3 receptor is beneficial and reduces cardiac dysfunction, inflammation, and HTN.NEW & NOTEWORTHY This study is the first to show that systemic treatment with an EP3 receptor antagonist (L798,106) attenuates the angiotensin II-induced increase in blood pressure in mice. The results from this project could complement existing hypertension therapies by combining blockade of the EP3 receptor with antihypertensive drugs.
Collapse
Affiliation(s)
- Timothy D Bryson
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan.,Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan
| | - Teja S Pandrangi
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan
| | - Safa Z Khan
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan
| | - Jiang Xu
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan
| | - Tengis S Pavlov
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan
| | - Pablo A Ortiz
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan.,Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan
| | - Edward Peterson
- Department of Public Health Sciences, Henry Ford Health System, Detroit, Michigan
| | - Pamela Harding
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan.,Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan
| |
Collapse
|
167
|
Ke M, Ji M, Wang H, Yao Y, Wu Y, Qi N. Inhibition of Rho-associated protein kinase improves the survival of human induced pluripotent stem cell-derived cardiomyocytes after dissociation. Exp Ther Med 2020; 19:1701-1710. [PMID: 32104223 PMCID: PMC7027158 DOI: 10.3892/etm.2020.8436] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 10/08/2019] [Indexed: 12/17/2022] Open
Abstract
Heart disease remains the leading cause of morbidity and mortality worldwide. Induced pluripotent stem cells (iPSCs) have the ability to differentiate into cardiomyocytes (CMs), rendering this cell type to be a promising pre-cursor of cardiomyocytes for cell-based cardiac regeneration. Obtaining CMs with a high yield and purity coupled with improved subsequent survival could prove to be invaluable for the future cell replacement therapeutic strategies. Rho-associated protein kinase (ROCK) is involved in a wide range of fundamental cellular functions and serves significant roles in cardiac physiology. In the present study, human (h)iPSC-CMs were generated from iPSCs by including glycogen synthase kinase 3β and Wnt inhibitors in the basal culture media. The possible effect of Y27632, a ROCK inhibitor, on hiPSC-CMs was then investigated. hiPSC-CMs of high purity were harvested with >96% of cells expressing cardiac troponin T. Additionally, treatment with 10 µM Y27632 significantly improved the viability of dissociated hiPSC-CMs. The effects of ROCK inhibitors Y27632 and fasudil, on the proliferation and apoptosis of hiPSC-CMs were also examined. Treatment with ROCK inhibitors markedly enhanced hiPSC-CM proliferation, by up to 2.5-fold, whilst Y27632 treatment reduced apoptosis in hiPSC-derived CMs under serum starvation and suspension by suppressing the expression of caspase-3. Taken together, data from the present study indicated that ROCK kinase inhibitors effectively improved the cultural system of hiPSC-derived CMs.
Collapse
Affiliation(s)
- Minxia Ke
- Department of Biochemistry and Molecular Biology, College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, P.R. China
| | - Meng Ji
- Hangzhou Biaomo Biosciences Co., Ltd., Hangzhou, Zhejiang 310018, P.R. China
| | - Hao Wang
- Shanghai Likun Biosciences Co., Ltd., Shanghai 201499, P.R. China
| | - Yifeng Yao
- Department of Biochemistry and Molecular Biology, College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, P.R. China
| | - Yuehong Wu
- Department of Biochemistry and Molecular Biology, College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, P.R. China
| | - Nianmin Qi
- Shanghai Likun Biosciences Co., Ltd., Shanghai 201499, P.R. China
| |
Collapse
|
168
|
Moodley S, Lian EY, Crupi MJF, Hyndman BD, Mulligan LM. RET isoform-specific interaction with scaffold protein Ezrin promotes cell migration and chemotaxis in lung adenocarcinoma. Lung Cancer 2020; 142:123-131. [PMID: 32146264 DOI: 10.1016/j.lungcan.2020.02.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/09/2020] [Accepted: 02/11/2020] [Indexed: 12/16/2022]
Abstract
OBJECTIVES Increased expression of REarranged during Transfection (RET) kinase is reported in 10-20 % of lung adenocarcinomas (LUAD) and is associated with metastasis and reduced survival. Ezrin is a scaffold protein that promotes protein interactions with the actin cytoskeleton to regulate cell migration and is also associated with invasion and metastasis in cancers. RET isoforms interact with unique combinations of scaffold proteins to promote distinct signaling pathways. We hypothesized that RET isoforms associate distinctly with Ezrin for cytoskeletal reorganization and LUAD cell migration processes. METHODS HCC1833 and A549 LUAD, SH-SY5Y neuroblastoma or HEK-293 cells expressing RET and Ezrin were stimulated with the RET ligand glial cell line-derived neurotrophic factor (GDNF) and treated with RET, Ezrin or Src inhibitors. Co-immunoprecipitation or pull-down assays coupled to immunoblotting were used to investigate protein activation and interactions. Immunofluorescence confocal microscopy assessed LUAD cytoskeletal reorganization and colocalization of RET and Ezrin. Live-cell fluorescence imaging was used to measure cell migration and chemotaxis. RESULTS GDNF promoted activation, interaction and colocalization of RET51 isoform and Ezrin. Inhibition of RET or Src impaired Ezrin interactions with RET and Src. GDNF stimulation enhanced the formation of actin-rich filopodia, in which both RET and Ezrin were enriched, and promoted chemotaxis in LUAD cells. However, inhibition of RET, Src or Ezrin suppressed filopodia formation, reduced colocalization of Ezrin with RET, and impaired cell migration and/ or chemotaxis. We further showed that GDNF-mediated activation of RET and Ezrin promoted RhoA-GTPase activity and signaling of ROCK1 and ROCK2 in LUAD cells. CONCLUSIONS Expression and activation of RET51 mediates unique protein interactions with Ezrin to promote LUAD cell chemotaxis for cancer cell dissemination, which may have implications in LUAD metastatic progression.
Collapse
Affiliation(s)
- Serisha Moodley
- Division of Cancer Biology and Genetics, Cancer Research Institute and Department of Pathology & Molecular Medicine, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Eric Y Lian
- Division of Cancer Biology and Genetics, Cancer Research Institute and Department of Pathology & Molecular Medicine, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Mathieu J F Crupi
- Division of Cancer Biology and Genetics, Cancer Research Institute and Department of Pathology & Molecular Medicine, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Brandy D Hyndman
- Division of Cancer Biology and Genetics, Cancer Research Institute and Department of Pathology & Molecular Medicine, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Lois M Mulligan
- Division of Cancer Biology and Genetics, Cancer Research Institute and Department of Pathology & Molecular Medicine, Queen's University, Kingston, ON, K7L 3N6, Canada.
| |
Collapse
|
169
|
Moretti R, Caruso P. Small Vessel Disease-Related Dementia: An Invalid Neurovascular Coupling? Int J Mol Sci 2020; 21:1095. [PMID: 32046035 PMCID: PMC7036993 DOI: 10.3390/ijms21031095] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 02/04/2020] [Accepted: 02/04/2020] [Indexed: 12/18/2022] Open
Abstract
The arteriosclerosis-dependent alteration of brain perfusion is one of the major determinants in small vessel disease, since small vessels have a pivotal role in the brain's autoregulation. Nevertheless, as far as we know, endothelium distress can potentiate the flow dysregulation and lead to subcortical vascular dementia that is related to small vessel disease (SVD), also being defined as subcortical vascular dementia (sVAD), as well as microglia activation, chronic hypoxia and hypoperfusion, vessel-tone dysregulation, altered astrocytes, and pericytes functioning blood-brain barrier disruption. The molecular basis of this pathology remains controversial. The apparent consequence (or a first event, too) is the macroscopic alteration of the neurovascular coupling. Here, we examined the possible mechanisms that lead a healthy aging process towards subcortical dementia. We remarked that SVD and white matter abnormalities related to age could be accelerated and potentiated by different vascular risk factors. Vascular function changes can be heavily influenced by genetic and epigenetic factors, which are, to the best of our knowledge, mostly unknown. Metabolic demands, active neurovascular coupling, correct glymphatic process, and adequate oxidative and inflammatory responses could be bulwarks in defense of the correct aging process; their impairments lead to a potentially catastrophic and non-reversible condition.
Collapse
Affiliation(s)
- Rita Moretti
- Neurology Clinic, Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy;
| | | |
Collapse
|
170
|
Enhanced cutaneous Rock2 expression as a marker of Rho Kinase pathway activation in autoimmune disease and Kohlemeier-Degos disease. Ann Diagn Pathol 2020; 44:151414. [DOI: 10.1016/j.anndiagpath.2019.151414] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 09/11/2019] [Indexed: 11/22/2022]
|
171
|
Gu Z, Yan T, Yan F. Rational design and improvement of the dimerization-disrupting peptide selectivity between ROCK-I and ROCK-II kinase isoforms in cerebrovascular diseases. J Mol Recognit 2020; 33:e2835. [PMID: 31995258 DOI: 10.1002/jmr.2835] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/17/2019] [Accepted: 12/25/2019] [Indexed: 02/05/2023]
Abstract
Human rho-associated coiled-coil forming kinases (ROCKs) ROCK-I and ROCK-II have been documented as attractive therapeutic targets for cerebrovascular diseases. Although ROCK-I and ROCK-II share a high degree of structural conservation and are both present in classic rho/ROCK signaling pathway, their downstream substrates and pathological functions may be quite different. Selective targeting of the two kinase isoforms with traditional small-molecule inhibitors is a great challenge due to their surprisingly high homology in kinase domain (~90%) and the full identity in kinase active site (100%). Here, instead of developing small-molecule drugs to selectively target the adenosine triphosphate (ATP) site of two isoforms, we attempt to design peptide agents to selectively disrupt the homo-dimerization event of ROCK kinases through their dimerization domains which have a relatively low conservation (~60%). Three helical peptides H1, H2, and H3 are split from the kinase dimerization domain, from which the isolated H2 peptide is found to have the best capability to rebind at the dimerization interface. A simulated annealing (SA) iteration method is used to improve the H2 peptide selectivity between ROCK-I and ROCK-II. The method accepts moderate degradation in peptide affinity in order to maximize the affinity difference between peptide binding to the two isoforms. Consequently, hundreds of parallel SA runs yielded six promising peptide candidates with ROCK-I over ROCK-II (I over II [IoII]) calculated selectivity and four promising peptide candidates with ROCK-II over ROCK-I (II over I [IIoI]) calculated selectivity. Subsequent anisotropy assays confirm that the selectivity values range between 13.2-fold and 83.9-fold for IoII peptides, and between 5.8-fold and 21.2-fold for IIoI peptides, which are considerably increased relative to wild-type H2 peptide (2.6-fold for IoII and 2.0-fold for IIoI). The molecular origin of the designed peptide selectivity is also analyzed at structural level; it is revealed that the peptide residues can be classified into conserved, non-conserved, and others, in which the non-conserved residues play a crucial role in defining peptide selectivity, while conserved residues confer stability to kinase-peptide binding.
Collapse
Affiliation(s)
- Zhengtian Gu
- Department of Neurology, Affiliated Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Tingting Yan
- Department of Pediatrics, Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| | - Fuling Yan
- Department of Neurology, Affiliated Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
172
|
Shahbazi R, Baradaran B, Khordadmehr M, Safaei S, Baghbanzadeh A, Jigari F, Ezzati H. Targeting ROCK signaling in health, malignant and non-malignant diseases. Immunol Lett 2020; 219:15-26. [PMID: 31904392 DOI: 10.1016/j.imlet.2019.12.012] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/15/2019] [Accepted: 12/30/2019] [Indexed: 12/21/2022]
Abstract
A Rho-associated coiled-coil kinase (ROCK) is identified as a critical downstream effector of GTPase RhoA which contains two isoforms, ROCK1 (also known as p160ROCK and ROKβ) and ROCK2 (also known as Rho-kinase and ROKα), the gene of which is placed on chromosomes 18 (18q11.1) and 2 (2p24), respectively. ROCKs have a principal function in the generation of actin-myosin contractility and regulation of actin cytoskeleton dynamics. They represent a chief role in regulating various cellular functions, such as apoptosis, growth, migration, and metabolism through modulation of cytoskeletal actin synthesis, and cellular contraction through phosphorylation of numerous downstream targets. Emerging evidence has indicated that ROCKs present a significant function in cardiac physiology. Of note, dysregulation of ROCKs involves in several cardiac pathological processes like cardiac hypertrophy, cardiac fibrosis, systemic blood pressure disorder, and pulmonary hypertension. Moreover, ROCKs, in addition to their role in regulating renal arteriolar contraction, glomerular blood flow, and filtration, can also play a role in controlling podocytes, tubular cells, and mesangial cell structure and function. Hyperactivity disorder and over-gene expression of Rho/ROCK have been indicated in different cancers. Furthermore, it seems that increasing the expression of mRNA or ROCK protein has an undesirable effect on patient survival and has a positive impact on the progression and worsening of disease prognosis. This review focuses on the physiological and pathological functions of ROCKs with a particular view on its possible value of ROCK inhibitors as a new therapy in cancers and non-cancer diseases.
Collapse
Affiliation(s)
- Roya Shahbazi
- Department of Pathology, Faculty of Veterinary Medicine, University of Tabriz, 51665-1647, Tabriz, Iran.
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, 51666-14761, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, 51666-14761, Tabriz, Iran.
| | - Monireh Khordadmehr
- Department of Pathology, Faculty of Veterinary Medicine, University of Tabriz, 51665-1647, Tabriz, Iran.
| | - Sahar Safaei
- Immunology Research Center, Tabriz University of Medical Sciences, 51666-14761, Tabriz, Iran.
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, 51666-14761, Tabriz, Iran.
| | - Farinaz Jigari
- Department of Pathology, Faculty of Veterinary Medicine, University of Tabriz, 51665-1647, Tabriz, Iran.
| | - Hamed Ezzati
- Department of Pathology, Faculty of Veterinary Medicine, University of Tabriz, 51665-1647, Tabriz, Iran.
| |
Collapse
|
173
|
Bober P, Alexovič M, Tomková Z, Kilík R, Sabo J. RHOA and mDia1 Promotes Apoptosis of Breast Cancer Cells Via a High Dose of Doxorubicin Treatment. Open Life Sci 2019; 14:619-627. [PMID: 33817200 PMCID: PMC7874778 DOI: 10.1515/biol-2019-0070] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 09/05/2019] [Indexed: 01/01/2023] Open
Abstract
Background Transforming RhoA proteins (RHOA) and their downstream Diaphanous homolog 1 proteins (DIAPH1) or mDia1 participate in the regulation of actin cytoskeleton which plays critical role in cells, i.e., morphologic changes and apoptosis. Methodology To determine the cell viability the real time cell analysis (RTCA) and flow cytometry were used. To perform proteomic analysis, the label-free quantitative method and post-translation modification by the nano-HPLC and ESI-MS ion trap mass analyser were used. Results The results of the cell viability showed an increase of dead cells (around 30 %) in MCF-7/DOX-1 (i.e., 1μM of doxorubicin was added to MCF-7/WT breast cancer cell line) compared to MCF-7/WT (control) after 24 h doxorubicin (DOX) treatment. The signalling pathway of the Regulation of actin cytoskeleton (p<0.0026) was determined, where RHOA and mDia1 proteins were up-regulated. Also, post-translational modification analysis of these proteins in MCF-7/DOX-1 cells revealed dysregulation of the actin cytoskeleton, specifically the collapse of actin stress fibbers due to phosphorylation of RHOA at serine 188 and mDia1 at serine 22, resulting in their deactivation and cell apoptosis. Conclusion These results pointed to an assumed role of DOX to dysregulation of actin cytoskeleton and cell death.
Collapse
Affiliation(s)
- Peter Bober
- Department of Medical and Clinical Biophysics, Faculty of Medicine, University of P.J. Šafárik in Košice, Trieda SNP1, 04011 Košice, Slovakia
| | - Michal Alexovič
- Department of Medical and Clinical Biophysics, Faculty of Medicine, University of P.J. Šafárik in Košice, Trieda SNP1, 04011 Košice, Slovakia
| | - Zuzana Tomková
- Department of Medical and Clinical Biophysics, Faculty of Medicine, University of P.J. Šafárik in Košice, Trieda SNP1, 04011 Košice, Slovakia
| | - Róbert Kilík
- 1st Department of Surgery, Faculty of Medicine, University of P.J. Šafárik in Košice, Trieda SNP1, 04011 Košice, Slovakia
| | - Ján Sabo
- Department of Medical and Clinical Biophysics, Faculty of Medicine, University of P.J. Šafárik in Košice, Trieda SNP1, 04011 Košice, Slovakia
| |
Collapse
|
174
|
Tello K, Seeger W, Naeije R, Vanderpool R, Ghofrani HA, Richter M, Tedford RJ, Bogaard HJ. Right heart failure in pulmonary hypertension: Diagnosis and new perspectives on vascular and direct right ventricular treatment. Br J Pharmacol 2019; 178:90-107. [PMID: 31517994 DOI: 10.1111/bph.14866] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 07/15/2019] [Accepted: 09/04/2019] [Indexed: 12/18/2022] Open
Abstract
Adaptation of right ventricular (RV) function to increased afterload-known as RV-arterial coupling-is a key determinant of prognosis in pulmonary hypertension. However, measurement of RV-arterial coupling is a complex, invasive process involving analysis of the RV pressure-volume relationship during preload reduction over multiple cardiac cycles. Simplified methods have therefore been proposed, including echocardiographic and cardiac MRI approaches. This review describes the available methods for assessment of RV function and RV-arterial coupling and the effects of pharmacotherapy on these variables. Overall, pharmacotherapies for pulmonary hypertension have shown beneficial effects on various measures of RV function, but it is often unclear if these are direct RV effects or indirect results of afterload reduction. Studies of the effects of pharmacotherapies on RV-arterial coupling are limited and mostly restricted to experimental models. Simplified methods to assess RV-arterial coupling should be validated and incorporated into routine clinical follow-up and future clinical trials. LINKED ARTICLES: This article is part of a themed issue on Risk factors, comorbidities, and comedications in cardioprotection. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.1/issuetoc.
Collapse
Affiliation(s)
- Khodr Tello
- Department of Internal Medicine, Justus-Liebig-University Giessen, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Giessen, Germany
| | - Werner Seeger
- Department of Internal Medicine, Justus-Liebig-University Giessen, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Giessen, Germany
| | - Robert Naeije
- Physiology, Erasme University Hospital, Brussels, Belgium
| | | | - Hossein Ardeschir Ghofrani
- Department of Internal Medicine, Justus-Liebig-University Giessen, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Giessen, Germany
| | - Manuel Richter
- Department of Internal Medicine, Justus-Liebig-University Giessen, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Giessen, Germany
| | - Ryan J Tedford
- Division of Cardiology, Department of Medicine, Medical University of South Carolina (MUSC), Charleston, SC, USA
| | - Harm J Bogaard
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
175
|
Ocaranza MP, Moya J, Jalil JE, Lavandero S, Kalergis AM, Molina C, Gabrielli L, Godoy I, Córdova S, Castro P, Mac Nab P, Rossel V, García L, González J, Mancilla C, Fierro C, Farías L. Rho-kinase pathway activation and apoptosis in circulating leucocytes in patients with heart failure with reduced ejection fraction. J Cell Mol Med 2019; 24:1413-1427. [PMID: 31778027 PMCID: PMC6991691 DOI: 10.1111/jcmm.14819] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 08/11/2019] [Accepted: 09/01/2019] [Indexed: 01/01/2023] Open
Abstract
Background Increased Rho‐kinase activity in circulating leucocytes is observed in heart failure with reduced ejection fraction (HFrEF). However, there is little information in HFrEF regarding other Rho‐kinase pathway components an on the relationship between Rho‐kinase and apoptosis. Here, Rho‐kinase activation levels and phosphorylation of major downstream molecules and apoptosis levels were measured for the first time both in HFrEF patients and healthy individuals. Methods Cross‐sectional study comparing HFrEF patients (n = 20) and healthy controls (n = 19). Rho‐kinase activity in circulating leucocytes (peripheral blood mononuclear cells, PBMCs) was determined by myosin light chain phosphatase 1 (MYPT1) and ezrin‐radixin‐moesin (ERM) phosphorylation. Rho‐kinase cascade proteins phosphorylation p38‐MAPK, myosin light chain‐2, JAK and JNK were also analysed along with apoptosis. Results MYPT1 and ERM phosphorylation were significantly elevated in HFrEF patients, (3.9‐ and 4.8‐fold higher than in controls, respectively). JAK phosphorylation was significantly increased by 300% over controls. Phosphorylation of downstream molecules p38‐MAPK and myosin light chain‐2 was significantly higher by 360% and 490%, respectively, while JNK phosphorylation was reduced by 60%. Catecholamine and angiotensin II levels were significantly higher in HFrEF patients, while angiotensin‐(1‐9) levels were lower. Apoptosis in circulating leucocytes was significantly increased in HFrEF patients by 2.8‐fold compared with controls and significantly correlated with Rho‐kinase activation. Conclusion Rho‐kinase pathway is activated in PMBCs from HFrEF patients despite optimal treatment, and it is closely associated with neurohormonal activation and with apoptosis. ROCK cascade inhibition might induce clinical benefits in HFrEF patients, and its assessment in PMBCs could be useful to evaluate reverse remodelling and disease regression.
Collapse
Affiliation(s)
- Maria Paz Ocaranza
- Department of Cardiovascular Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jackeline Moya
- Department of Cardiovascular Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jorge E Jalil
- Department of Cardiovascular Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Sergio Lavandero
- Faculty of Chemical and Pharmaceutical Sciences, Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santiago, Chile.,Cardiology Division, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Alexis M Kalergis
- Departament of Molecular Genetics and Microbiology, Faculty of Biological Sciences, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cristián Molina
- Department of Cardiovascular Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Luigi Gabrielli
- Department of Cardiovascular Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Iván Godoy
- Department of Cardiovascular Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Samuel Córdova
- Department of Cardiovascular Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo Castro
- Department of Cardiovascular Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Paul Mac Nab
- Department of Cardiovascular Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Victor Rossel
- Department of Medicine, Hospital del Salvador, Medical School, Universidad de Chile, Santiago, Chile
| | - Lorena García
- Faculty of Chemical and Pharmaceutical Sciences, Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santiago, Chile
| | - Javier González
- Department of Cardiovascular Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cristián Mancilla
- Department of Cardiovascular Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Camila Fierro
- Department of Cardiovascular Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Luis Farías
- Department of Cardiovascular Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
176
|
Nimgampalle M, Banavath HN, Chakravarthy H, Saxena A, Devanathan V. Novel inhibitors of Rho-kinase mediated neuroinflammatory pathways and their potential application in recovery of injured spinal cord. J Biomol Struct Dyn 2019; 38:4669-4686. [DOI: 10.1080/07391102.2019.1686066] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Mallikarjuna Nimgampalle
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, Andhra Pradesh, India
| | - Hemanth Naick Banavath
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, Andhra Pradesh, India
| | - Harshini Chakravarthy
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, Andhra Pradesh, India
| | | | - Vasudharani Devanathan
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, Andhra Pradesh, India
| |
Collapse
|
177
|
Klepac K, Yang J, Hildebrand S, Pfeifer A. RGS2: A multifunctional signaling hub that balances brown adipose tissue function and differentiation. Mol Metab 2019; 30:173-183. [PMID: 31767169 PMCID: PMC6807268 DOI: 10.1016/j.molmet.2019.09.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/12/2019] [Accepted: 09/28/2019] [Indexed: 12/28/2022] Open
Abstract
Objective Recruitment of brown adipose tissue (BAT) is a potential new strategy for increasing energy expenditure (EE) to treat obesity. G protein–coupled receptors (GPCRs) represent promising targets to activate BAT, as they are the major regulators of BAT biological function. To identify new regulators of GPCR signaling in BAT, we studied the role of Regulator of G protein Signaling 2 (RGS2) in brown adipocytes and BAT. Methods We combined pharmacological and genetic tools to investigate the role of RGS2 in BAT in vitro and in vivo. Adipocyte progenitors were isolated from wild-type (WT) and RGS2 knockout (RGS2−/−) BAT and differentiated to brown adipocytes. This approach was complemented with knockdown of RGS2 using lentiviral shRNAs (shRGS2). Adipogenesis was analyzed by Oil Red O staining and by determining the expression of adipogenic and thermogenic markers. Pharmacological modulators and fluorescence staining of F-acting stress fibers were employed to identify the underlying signaling pathways. In vivo, the activity of BAT was assessed by ex vivo lipolysis and by measuring whole-body EE by indirect calorimetry in metabolic cages. Results RGS2 is highly expressed in BAT, and treatment with cGMP—an important enhancer of brown adipocyte differentiation—further increased RGS2 expression. Loss of RGS2 strongly suppressed adipogenesis and the expression of thermogenic genes in brown adipocytes. Mechanistically, we found increased Gq/Rho/Rho kinase (ROCK) signaling in the absence of RGS2. Surprisingly, in vivo analysis revealed elevated BAT activity in RGS2-deficient mice that was caused by enhanced Gs/cAMP signaling. Conclusion Overall, RGS2 regulates two major signaling pathways in BAT: Gq and Gs. On the one hand, RGS2 promotes brown adipogenesis by counteracting the inhibitory action of Gq/Rho/ROCK signaling. On the other hand, RGS2 decreases the activity of BAT through the inhibition of Gs signaling and cAMP production. Thus, RGS2 might represent a stress modulator that protects BAT from overstimulation. RGS2 regulates brown adipose tissue (BAT) by inhibiting two major G protein-coupled receptor (GPCR) pathways – Gq and Gs. Deletion of RGS2 impairs the differentiation of murine brown adipocytes due to elevated Gq/Rho/ROCK signaling. In vivo, RGS2 knock-out mice show an increase in BAT lipolysis and whole-body energy expenditure.
Collapse
Affiliation(s)
- Katarina Klepac
- Institute of Pharmacology and Toxicology, University of Bonn, 53127 Bonn, Germany; Research Training Group 1873, University of Bonn, 53127 Bonn, Germany.
| | - JuHee Yang
- Institute of Pharmacology and Toxicology, University of Bonn, 53127 Bonn, Germany; Research Training Group 1873, University of Bonn, 53127 Bonn, Germany
| | - Staffan Hildebrand
- Institute of Pharmacology and Toxicology, University of Bonn, 53127 Bonn, Germany
| | - Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University of Bonn, 53127 Bonn, Germany; Research Training Group 1873, University of Bonn, 53127 Bonn, Germany; PharmaCenter, University of Bonn, 53127 Bonn, Germany.
| |
Collapse
|
178
|
Deng Z, Jia Y, Liu H, He M, Yang Y, Xiao W, Li Y. RhoA/ROCK pathway: implication in osteoarthritis and therapeutic targets. Am J Transl Res 2019; 11:5324-5331. [PMID: 31632513 PMCID: PMC6789288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 08/18/2019] [Indexed: 06/10/2023]
Abstract
Ras homolog gene family, member A (RhoA) and its downstream effector Rho-associated protein kinase (ROCK) play important roles in multiple cellular processes, but abnormal activation of this pathway have been reported to be involved in various types of diseases, including osteoarthritis (OA). This article focused to review the RhoA/ROCK association and its functional role in OA development, and possible therapeutics of OA by targeting this pathway. We have explored the databases like Pubmed, Google Scholar, Web of Science and SCOPUS, and collected the papers on Rho/ROCK and their relationship with OA, and reviewed comprehensively. Studies revealed that the abnormal activation of RhoA/ROCK signaling is involved in early phase response to abnormal mechanical stimuli, which is thought to be a contributory factor to OA progression. RhoA/ROCK interacts with OA pathological factors and induces cartilage degeneration through the degradation of chondrocyte extracellular matrix (ECM). As the RhoA/ROCK activity can affect bone formation by triggering cartilage degradation, it may represent a possible therapeutic target to treat OA. Interestingly, several pharmaceutical companies are investing in the development of RhoA/ROCK inhibitors for the treatment of OA. However, a few in vivo experiments have been successfully conducted to demonstrate the potential value of RhoA/ROCK pathway inhibition in the treatment of OA. This review provides an insight into the functional role of Rho/ROCK pathway, and indicates that targeting this pathway might be promising in future OA treatment.
Collapse
Affiliation(s)
- Zhenhan Deng
- Department of Orthopedics, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, China
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s HospitalShenzhen 518035, Guangdong, China
| | - Yiming Jia
- Department of Orthopedics, Chifeng Municipal Hospital, Chifeng Clinical Medical School of Inner Mongolia Medical UniversityChifeng 024000, Inner Mongolia, China
| | - Haifeng Liu
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s HospitalShenzhen 518035, Guangdong, China
| | - Miao He
- Department of Orthopedics, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, China
| | - Yuntao Yang
- Department of Orthopedics, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, China
| | - Wenfeng Xiao
- Department of Orthopedics, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, China
| |
Collapse
|
179
|
Stam K, Cai Z, van der Velde N, van Duin R, Lam E, van der Velden J, Hirsch A, Duncker DJ, Merkus D. Cardiac remodelling in a swine model of chronic thromboembolic pulmonary hypertension: comparison of right vs. left ventricle. J Physiol 2019; 597:4465-4480. [PMID: 31194256 PMCID: PMC6852085 DOI: 10.1113/jp277896] [Citation(s) in RCA: 14] [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: 02/25/2019] [Accepted: 06/07/2019] [Indexed: 12/11/2022] Open
Abstract
KEY POINTS Right ventricle (RV) function is the most important determinant of survival and quality of life in patients with chronic thromboembolic pulmonary hypertension (CTEPH). The changes in right and left ventricle gene expression that contribute to ventricular remodelling are incompletely investigated. RV remodelling in our CTEPH swine model is associated with increased expression of the genes involved in inflammation (TGFβ), oxidative stress (ROCK2, NOX1 and NOX4), and apoptosis (BCL2 and caspase-3). Alterations in ROCK2 expression correlated inversely with RV contractile reserve during exercise. Since ROCK2 has been shown to be involved in hypertrophy, oxidative stress, fibrosis and endothelial dysfunction, ROCK2 inhibition may present a viable therapeutic target in CTEPH. ABSTRACT Right ventricle (RV) function is the most important determinant of survival and quality of life in patients with chronic thromboembolic pulmonary hypertension (CTEPH). The present study investigated whether the increased cardiac afterload is associated with (i) cardiac remodelling and hypertrophic signalling; (ii) changes in angiogenic factors and capillary density; and (iii) inflammatory changes associated with oxidative stress and interstitial fibrosis. CTEPH was induced in eight chronically instrumented swine by chronic nitric oxide synthase inhibition and up to five weekly pulmonary embolizations. Nine healthy swine served as a control. After 9 weeks, RV function was assessed by single beat analysis of RV-pulmonary artery (PA) coupling at rest and during exercise, as well as by cardiac magnetic resonance imaging. Subsequently, the heart was excised and RV and left ventricle (LV) tissues were processed for molecular and histological analyses. Swine with CTEPH exhibited significant RV hypertrophy in response to the elevated PA pressure. RV-PA coupling was significantly reduced, correlated inversely with pulmonary vascular resistance and did not increase during exercise in CTEPH swine. Expression of genes associated with hypertrophy (BNP), inflammation (TGFβ), oxidative stress (ROCK2, NOX1 and NOX4), apoptosis (BCL2 and caspase-3) and angiogenesis (VEGFA) were increased in the RV of CTEPH swine and correlated inversely with RV-PA coupling during exercise. In the LV, only significant changes in ROCK2 gene-expression occurred. In conclusion, RV remodelling in our CTEPH swine model is associated with increased expression of genes involved in inflammation and oxidative stress, suggesting that these processes contribute to RV remodelling and dysfunction in CTEPH and hence represent potential therapeutic targets.
Collapse
Affiliation(s)
- Kelly Stam
- Department of Cardiology, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Zongye Cai
- Department of Cardiology, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Nikki van der Velde
- Department of Cardiology, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Richard van Duin
- Department of Cardiology, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Esther Lam
- Department of Cardiology, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Jolanda van der Velden
- Amsterdam UMCVrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular SciencesAmsterdamThe Netherlands
| | - Alexander Hirsch
- Department of Cardiology, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Dirk J Duncker
- Department of Cardiology, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Daphne Merkus
- Department of Cardiology, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| |
Collapse
|
180
|
Santos GL, Hartmann S, Zimmermann WH, Ridley A, Lutz S. Inhibition of Rho-associated kinases suppresses cardiac myofibroblast function in engineered connective and heart muscle tissues. J Mol Cell Cardiol 2019; 134:13-28. [DOI: 10.1016/j.yjmcc.2019.06.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 06/01/2019] [Accepted: 06/20/2019] [Indexed: 12/13/2022]
|
181
|
Peng Z, Li X, Fu M, Zhu K, Long L, Zhao X, Chen Q, Deng DYB, Wan Y. Inhibition of Notch1 signaling promotes neuronal differentiation and improves functional recovery in spinal cord injury through suppressing the activation of Ras homolog family member A. J Neurochem 2019; 150:709-722. [PMID: 31339573 DOI: 10.1111/jnc.14833] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Accepted: 06/17/2019] [Indexed: 12/28/2022]
Abstract
Neural stem cells (NSCs) transplantation represents a promising strategy for the repair of injured neurons, since NSCs not only produce multiple neurotrophic growth factors but also differentiate into mature cells to replace damaged cells. Previous studies have shown that Notch signaling pathway had negative effects on neuronal differentiation; however, the precise mechanism remained inadequately understood. This research aimed to investigate whether inhibition of Notch1 signaling promotes neuronal differentiation and improves functional recovery in rat spinal cord injury through suppressing the activation of Ras homolog family member A (RhoA). QPCR, western blot, and immunofluorescence experiments were used to analyze Notch1 signaling pathways, RhoA, Ras homologous -associated coiled-coil containing protein kinase 1 (ROCK1), cleaved caspased-3, and neuronal/astrocytic differentiation markers. The expression of RhoA and ROCK1 was inhibited by lentivirus or specific biochemical inhibitors. In spinal cord injury (SCI), motor function was assessed by hind limbs movements and electrophysiology. Tissue repairing was measured by immunofluorescence, Nissl staining, Fluorogold, HE staining, QPCR, western blot, and magnetic resonance imaging (MRI) experiments. Our results demonstrate that inhibition of Notch1 in NSCs can promote the differentiation of NSCs to neurons. Knockdown of RhoA and inhibition of ROCK1 both can promote neuronal differentiation through inhibiting the activation of Notch1 signaling pathway in NSCs. In SCI, silencing RhoA enhanced neuronal differentiation and improved tissue repairing/functional recovery by inhibiting the activation of Notch1 signaling pathway. Since Notch1 inhibits neuronal differentiation through activating the RhoA/ROCK1 signaling pathway in NSCs, our data suggest that the Notch1/RhoA/ROCK1/Hes1/Hes5 signaling pathway may serve as a novel target for the treatment of SCI.
Collapse
Affiliation(s)
- Zhiming Peng
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiang Li
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Mengxia Fu
- Division of Cardiac Surgery, NHC Key Laboratory of Assisted Circulation, Ministry of Health, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kai Zhu
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lingli Long
- Department of Translational Medicine Center Research Laboratory, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoyang Zhao
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qingui Chen
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - David Y B Deng
- Scientific Research Center and Department of Orthopedic, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yong Wan
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
182
|
McKerracher L, Shenkar R, Abbinanti M, Cao Y, Peiper A, Liao JK, Lightle R, Moore T, Hobson N, Gallione C, Ruschel J, Koskimäki J, Girard R, Rosen K, Marchuk DA, Awad IA. A Brain-Targeted Orally Available ROCK2 Inhibitor Benefits Mild and Aggressive Cavernous Angioma Disease. Transl Stroke Res 2019; 11:365-376. [PMID: 31446620 DOI: 10.1007/s12975-019-00725-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 08/01/2019] [Accepted: 08/13/2019] [Indexed: 12/01/2022]
Abstract
Cavernous angioma (CA) is a vascular pathology caused by loss of function in one of the 3 CA genes (CCM1, CCM2, and CCM3) that result in rho kinase (ROCK) activation. We investigated a novel ROCK2 selective inhibitor for the ability to reduce brain lesion formation, growth, and maturation. We used genetic methods to explore the use of a ROCK2-selective kinase inhibitor to reduce growth and hemorrhage of CAs. The role of ROCK2 in CA was investigated by crossing Rock1 or Rock2 hemizygous mice with Ccm1 or Ccm3 hemizygous mice, and we found reduced lesions in the Rock2 hemizygous mice. A ROCK2-selective inhibitor, BA-1049 was used to investigate efficacy in reducing CA lesions after oral administration to Ccm1+/- and Ccm3+/- mice that were bred into a mutator background. After assessing the dose range effective to target brain endothelial cells in an ischemic brain model, Ccm1+/- and Ccm3+/- transgenic mice were treated for 3 (Ccm3+/-) or 4 months (Ccm1+/-), concurrently, randomized to receive one of three doses of BA-1049 in drinking water, or placebo. Lesion volumes were assessed by micro-computed tomography. BA-1049 reduced activation of ROCK2 in Ccm3+/-Trp53-/- lesions. Ccm1+/-Msh2-/- (n=68) and Ccm3+/-Trp53-/- (n=71) mice treated with BA-1049 or placebo showed a significant dose-dependent reduction in lesion volume after treatment with BA-1049, and a reduction in hemorrhage (iron deposition) near lesions at all doses. These translational studies show that BA-1049 is a promising therapeutic agent for the treatment of CA, a disease with no current treatment except surgical removal of the brain lesions.
Collapse
Affiliation(s)
- Lisa McKerracher
- BioAxone BioSciences Inc., Cambridge, MA, USA.,Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
| | - Robert Shenkar
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, The University of Chicago Medicine, Chicago, IL, USA
| | | | - Ying Cao
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, The University of Chicago Medicine, Chicago, IL, USA
| | - Amy Peiper
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | - James K Liao
- Section of Cardiology, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Rhonda Lightle
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, The University of Chicago Medicine, Chicago, IL, USA
| | - Thomas Moore
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, The University of Chicago Medicine, Chicago, IL, USA
| | - Nicholas Hobson
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, The University of Chicago Medicine, Chicago, IL, USA
| | - Carol Gallione
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | | | - Janne Koskimäki
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, The University of Chicago Medicine, Chicago, IL, USA
| | - Romuald Girard
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, The University of Chicago Medicine, Chicago, IL, USA
| | | | - Douglas A Marchuk
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | - Issam A Awad
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, The University of Chicago Medicine, Chicago, IL, USA.
| |
Collapse
|
183
|
Butler MB, Short NE, Maniou E, Alexandre P, Greene NDE, Copp AJ, Galea GL. Rho kinase-dependent apical constriction counteracts M-phase apical expansion to enable mouse neural tube closure. J Cell Sci 2019; 132:jcs.230300. [PMID: 31182644 PMCID: PMC6633395 DOI: 10.1242/jcs.230300] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 05/30/2019] [Indexed: 12/18/2022] Open
Abstract
Cellular generation of mechanical forces required to close the presumptive spinal neural tube, the 'posterior neuropore' (PNP), involves interkinetic nuclear migration (INM) and apical constriction. Both processes change the apical surface area of neuroepithelial cells, but how they are biomechanically integrated is unknown. Rho kinase (Rock; herein referring to both ROCK1 and ROCK2) inhibition in mouse whole embryo culture progressively widens the PNP. PNP widening is not caused by increased mechanical tension opposing closure, as evidenced by diminished recoil following laser ablation. Rather, Rock inhibition diminishes neuroepithelial apical constriction, producing increased apical areas in neuroepithelial cells despite diminished tension. Neuroepithelial apices are also dynamically related to INM progression, with the smallest dimensions achieved in cells positive for the pan-M phase marker Rb phosphorylated at S780 (pRB-S780). A brief (2 h) Rock inhibition selectively increases the apical area of pRB-S780-positive cells, but not pre-anaphase cells positive for phosphorylated histone 3 (pHH3+). Longer inhibition (8 h, more than one cell cycle) increases apical areas in pHH3+ cells, suggesting cell cycle-dependent accumulation of cells with larger apical surfaces during PNP widening. Consequently, arresting cell cycle progression with hydroxyurea prevents PNP widening following Rock inhibition. Thus, Rock-dependent apical constriction compensates for the PNP-widening effects of INM to enable progression of closure.This article has an associated First Person interview with the first authors of the paper.
Collapse
Affiliation(s)
- Max B Butler
- Developmental Biology and Cancer, UCL GOS Institute of Child Health, London WC1N 1EH, UK
| | - Nina E Short
- Developmental Biology and Cancer, UCL GOS Institute of Child Health, London WC1N 1EH, UK
| | - Eirini Maniou
- Developmental Biology and Cancer, UCL GOS Institute of Child Health, London WC1N 1EH, UK
| | - Paula Alexandre
- Developmental Biology and Cancer, UCL GOS Institute of Child Health, London WC1N 1EH, UK
| | - Nicholas D E Greene
- Developmental Biology and Cancer, UCL GOS Institute of Child Health, London WC1N 1EH, UK
| | - Andrew J Copp
- Developmental Biology and Cancer, UCL GOS Institute of Child Health, London WC1N 1EH, UK
| | - Gabriel L Galea
- Developmental Biology and Cancer, UCL GOS Institute of Child Health, London WC1N 1EH, UK .,Comparative Bioveterinary Sciences, Royal Veterinary College, London NW1 0TU, UK
| |
Collapse
|
184
|
Paula SM, Couto GK, Fontes MT, Costa SK, Negrão CE, Mill JG, Rossoni LV. Exercise training restores the myogenic response in skeletal muscle resistance arteries and corrects peripheral edema in rats with heart failure. Am J Physiol Heart Circ Physiol 2019; 317:H87-H96. [DOI: 10.1152/ajpheart.00042.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Impairment of the myogenic response can affect capillary hydrostatic pressure and contribute to peripheral edema and exercise intolerance, which are markers of heart failure (HF). The aim of this study was to assess the effects of exercise training (ET) on myogenic response in skeletal muscle resistance arteries and peripheral edema in HF rats, focusing on the potential signaling pathways involved in these adjustments. Male Wistar rats were submitted to either coronary artery occlusion or a sham-operated surgery. After 4 wk, an exercise test was performed, and the rats were divided into the following groups: untrained normal control (UNC) and untrained HF (UHF) and exercise- trained (on treadmill, 50–60% of maximal capacity) NC (TNC) and exercise-trained HF (THF). Caudal tibial artery (CTA) myogenic response was impaired in UHF compared with UNC, and ET restored this response in THF to NC levels and increased it in TNC. Rho kinase (ROCK) inhibitor abolished CTA myogenic response in the untrained and blunted it in exercise-trained groups. CTA-stored calcium (Ca2+) mobilization was higher in exercise-trained rats compared with untrained rats. The paw volume was higher in UHF rats, and ET decreased this response compared with UNC. Myogenic constriction was positively correlated with maximal running distance and negatively correlated with paw volume. The results demonstrate, for the first time, that HF impairs the myogenic response in skeletal muscle arteries, which contributes to peripheral edema in this syndrome. ET restores the myogenic response in skeletal muscle arteries improving Ca2+ sensitization and handling. Additionally, this paradigm also improves peripheral edema and exercise intolerance. NEW & NOTEWORTHY The novel and main finding of the present study is that moderate intensity exercise training restores the impaired myogenic response of skeletal muscle resistance arteries, exercise intolerance and peripheral edema in rats with heart failure. These results also show for the first time to our knowledge that exercise training improving calcium sensitization through the ROCK pathway and enhancing intracellular calcium handling could contribute to restoration of flow autoregulation to skeletal muscle in heart failure.
Collapse
Affiliation(s)
- Suliana M. Paula
- Department of Physiology and Biophysics, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Gisele K. Couto
- Department of Physiology and Biophysics, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Milene T. Fontes
- Department of Physiology and Biophysics, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Soraia K. Costa
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Carlos E. Negrão
- Heart Institute, Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, University of São Paulo, São Paulo, Brazil
- School of Physical Education and Sport, University of São Paulo, São Paulo, Brazil
| | - José G. Mill
- Department of Physiological Sciences, Federal University of Espírito Santo, Espírito Santo, Brazil
| | - Luciana V. Rossoni
- Department of Physiology and Biophysics, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
185
|
Liu WY, Tang Q, Zhang Q, Hu CP, Huang JB, Sheng FF, Liu YL, Zhou M, Lai WJ, Li GB, Zhang R. Lycorine Induces Mitochondria-Dependent Apoptosis in Hepatoblastoma HepG2 Cells Through ROCK1 Activation. Front Pharmacol 2019; 10:651. [PMID: 31263414 PMCID: PMC6589644 DOI: 10.3389/fphar.2019.00651] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 05/20/2019] [Indexed: 01/13/2023] Open
Abstract
Lycorine, a naturally occurring compound extracted from the Amaryllidaceae plant family, has been reported to exhibit antitumor activity in various cancer cell types. In the present study, we investigated the molecular mechanisms underlying lycorine-induced apoptosis in hepatoblastoma HepG2 cells. We found that lycorine induced mitochondria-dependent apoptosis in HepG2 cells accompanied by mitochondrial permeability transition pore (mPTP) opening, mitochondrial membrane potential (MMP) loss, adenosine triphosphate (ATP) depletion, Ca2+ and cytochrome c (Cyto C) release, as well as caspase activation. Furthermore, we found Rho associated coiled-coil containing protein kinase 1 (ROCK1) cleavage/activation played a critical role in lycorine-induced mitochondrial apoptosis. In addition, the ROCK inhibitor Y-27632 was employed, and we found that co-treatment with Y-27632 attenuated lycorine-induced mitochondrial injury and cell apoptosis. Meanwhile, an in vivo study revealed that lycorine inhibited tumor growth and induced apoptosis in a HepG2 xenograft mouse model in association with ROCK1 activation. Taken together, all these findings suggested that lycorine induced mitochondria-dependent apoptosis through ROCK1 activation in HepG2 cells, and this may be a theoretical basis for lycorine's anticancer effects.
Collapse
Affiliation(s)
- Wu-Yi Liu
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Qin Tang
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Qian Zhang
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Chang-Peng Hu
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Jing-Bin Huang
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Fang-Fang Sheng
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Ya-Li Liu
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Min Zhou
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Wen-Jing Lai
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Guo-Bing Li
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Rong Zhang
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
186
|
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
|
187
|
Wang J, Sui RX, Miao Q, Wang Q, Song LJ, Yu JZ, Li YH, Xiao BG, Ma CG. Effect of Fasudil on remyelination following cuprizone-induced demyelination. CNS Neurosci Ther 2019; 26:76-89. [PMID: 31124292 PMCID: PMC6930827 DOI: 10.1111/cns.13154] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 04/29/2019] [Accepted: 05/07/2019] [Indexed: 01/24/2023] Open
Abstract
Background Multiple sclerosis is characterized by demyelination/remyelination, neuroinflammation, and neurodegeneration. Cuprizone (CPZ)‐induced toxic demyelination is an experimental animal model commonly used to study demyelination and remyelination in the central nervous system. Fasudil is one of the most thoroughly studied Rho kinase inhibitors. Methods Following CPZ exposure, the degree of demyelination in the brain of male C57BL/6 mice was assessed by Luxol fast blue, Black Gold II, myelin basic protein immunofluorescent staining, and Western blot. The effect of Fasudil on behavioral change was determined using elevated plus maze test and pole test. The possible mechanisms of Fasudil action were examined by immunohistochemistry, flow cytometry, ELISA, and dot blot. Results Fasudil improved behavioral abnormalities, inhibited microglia‐mediated neuroinflammation, and promoted astrocyte‐derived nerve growth factor and ciliary neurotrophic factor, which should contribute to protection and regeneration of oligodendrocytes. In addition, Fasudil inhibited the production of myelin oligodendrocyte glycoprotein antibody and the infiltration of peripheral CD4+ T cells and CD68+ macrophages, which appears to be related to the integrity of the blood‐brain barrier. Conclusion These results provide evidence for the therapeutic potential of Fasudil in CPZ‐induced demyelination. However, how Fasudil acts on microglia, astrocytes, and immune cells remains to be further explored.
Collapse
Affiliation(s)
- Jing Wang
- Department of Neurology, First Affiliated Hospital, Shanxi Medical University, Taiyuan, China
| | - Ruo-Xuan Sui
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Shanxi University of Traditional Chinese Medicine, Taiyuan, China
| | - Qiang Miao
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Shanxi University of Traditional Chinese Medicine, Taiyuan, China
| | - Qing Wang
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Shanxi University of Traditional Chinese Medicine, Taiyuan, China
| | - Li-Juan Song
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Shanxi University of Traditional Chinese Medicine, Taiyuan, China
| | - Jie-Zhong Yu
- Institute of Brain Science, Shanxi Datong University, Datong, China
| | - Yan-Hua Li
- Institute of Brain Science, Shanxi Datong University, Datong, China
| | - Bao-Guo Xiao
- Institute of Neurology, Huashan Hospital, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Cun-Gen Ma
- Department of Neurology, First Affiliated Hospital, Shanxi Medical University, Taiyuan, China.,The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Shanxi University of Traditional Chinese Medicine, Taiyuan, China.,Institute of Brain Science, Shanxi Datong University, Datong, China
| |
Collapse
|
188
|
Nair AR, Silva SD, Agbor LN, Wu J, Nakagawa P, Mukohda M, Lu KT, Sandgren JA, Pierce GL, Santillan MK, Grobe JL, Sigmund CD. Endothelial PPARγ (Peroxisome Proliferator-Activated Receptor-γ) Protects From Angiotensin II-Induced Endothelial Dysfunction in Adult Offspring Born From Pregnancies Complicated by Hypertension. Hypertension 2019; 74:173-183. [PMID: 31104564 DOI: 10.1161/hypertensionaha.119.13101] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Preeclampsia is a hypertensive disorder of pregnancy associated with vascular dysfunction and cardiovascular risk to offspring. We hypothesize that endothelial PPARγ (peroxisome proliferator-activated receptor-γ) provides cardiovascular protection in offspring from pregnancies complicated by hypertension. C57BL/6J dams were bred with E-V290M sires, which express a dominant-negative allele of PPARγ selectively in the endothelium. Arginine vasopressin was infused throughout gestation. Vasopressin elevated maternal blood pressure at gestational day 14 to 15 and urinary protein at day 17 consistent. Systolic blood pressure and vasodilation responses to acetylcholine were similar in vasopressin-exposed offspring compared to offspring from control pregnancies. We treated offspring with a subpressor dose of angiotensin II to test if hypertension during pregnancy predisposes offspring to hypertension. Male and female angiotensin II-treated E-V290M offspring from vasopressin-exposed but not control pregnancy exhibited significant impairment in acetylcholine-induced relaxation in carotid artery. Endothelial dysfunction in angiotensin II-treated E-V290M vasopressin-exposed offspring was attenuated by tempol, an effect which was more prominent in male offspring. Nrf2 (nuclear factor-E2-related factor) protein levels were significantly elevated in aorta from male E-V290M offspring, but not female offspring compared to controls. Blockade of ROCK (Rho-kinase) signaling and incubation with a ROCK2-specific inhibitor improved endothelial function in both male and female E-V290M offspring from vasopressin-exposed pregnancy. Our data suggest that interference with endothelial PPARγ in offspring from vasopressin-exposed pregnancies increases the risk for endothelial dysfunction on exposure to a cardiovascular stressor in adulthood. This implies that endothelial PPARγ provides protection to cardiovascular stressors in offspring of a pregnancy complicated by hypertension and perhaps in preeclampsia.
Collapse
Affiliation(s)
- Anand R Nair
- From the Department of Pharmacology (A.R.N., S.D.S., L.N.A., J.W., P.N., M.M., K.-T.L., J.A.S., J.L.G., C.D.S.), Roy J. and Lucille A. Carver College of Medicine, University of Iowa
| | - Sebastiao D Silva
- From the Department of Pharmacology (A.R.N., S.D.S., L.N.A., J.W., P.N., M.M., K.-T.L., J.A.S., J.L.G., C.D.S.), Roy J. and Lucille A. Carver College of Medicine, University of Iowa.,Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee (S.D.S., J.W., P.N., K.-T.L., J.L.G., C.D.S.)
| | - Larry N Agbor
- From the Department of Pharmacology (A.R.N., S.D.S., L.N.A., J.W., P.N., M.M., K.-T.L., J.A.S., J.L.G., C.D.S.), Roy J. and Lucille A. Carver College of Medicine, University of Iowa
| | - Jing Wu
- From the Department of Pharmacology (A.R.N., S.D.S., L.N.A., J.W., P.N., M.M., K.-T.L., J.A.S., J.L.G., C.D.S.), Roy J. and Lucille A. Carver College of Medicine, University of Iowa.,Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee (S.D.S., J.W., P.N., K.-T.L., J.L.G., C.D.S.)
| | - Pablo Nakagawa
- From the Department of Pharmacology (A.R.N., S.D.S., L.N.A., J.W., P.N., M.M., K.-T.L., J.A.S., J.L.G., C.D.S.), Roy J. and Lucille A. Carver College of Medicine, University of Iowa
| | - Masashi Mukohda
- From the Department of Pharmacology (A.R.N., S.D.S., L.N.A., J.W., P.N., M.M., K.-T.L., J.A.S., J.L.G., C.D.S.), Roy J. and Lucille A. Carver College of Medicine, University of Iowa
| | - Ko-Ting Lu
- From the Department of Pharmacology (A.R.N., S.D.S., L.N.A., J.W., P.N., M.M., K.-T.L., J.A.S., J.L.G., C.D.S.), Roy J. and Lucille A. Carver College of Medicine, University of Iowa.,Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee (S.D.S., J.W., P.N., K.-T.L., J.L.G., C.D.S.)
| | - Jeremy A Sandgren
- From the Department of Pharmacology (A.R.N., S.D.S., L.N.A., J.W., P.N., M.M., K.-T.L., J.A.S., J.L.G., C.D.S.), Roy J. and Lucille A. Carver College of Medicine, University of Iowa
| | - Gary L Pierce
- Department of Health and Human Physiology (G.L.P.), Roy J. and Lucille A. Carver College of Medicine, University of Iowa
| | - Mark K Santillan
- Department of Obstetrics and Gynecology (M.K.S.), Roy J. and Lucille A. Carver College of Medicine, University of Iowa
| | - Justin L Grobe
- From the Department of Pharmacology (A.R.N., S.D.S., L.N.A., J.W., P.N., M.M., K.-T.L., J.A.S., J.L.G., C.D.S.), Roy J. and Lucille A. Carver College of Medicine, University of Iowa.,Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee (S.D.S., J.W., P.N., K.-T.L., J.L.G., C.D.S.)
| | - Curt D Sigmund
- From the Department of Pharmacology (A.R.N., S.D.S., L.N.A., J.W., P.N., M.M., K.-T.L., J.A.S., J.L.G., C.D.S.), Roy J. and Lucille A. Carver College of Medicine, University of Iowa.,Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee (S.D.S., J.W., P.N., K.-T.L., J.L.G., C.D.S.)
| |
Collapse
|
189
|
Zhang L, Zhou H, Wei G. miR-506 regulates cell proliferation and apoptosis by affecting RhoA/ROCK signaling pathway in hepatocellular carcinoma cells. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:1163-1173. [PMID: 31933931 PMCID: PMC6947048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 12/26/2018] [Indexed: 06/10/2023]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC), is the third leading cause of cancer-related death. MicroRNA-506 (miR-506) has been reported to exhibit abnormal expression in HCC; however, the role of miR-506 in HCC and the molecular mechanisms underlying miR-506 in HCC remain unclarified. METHODS Quantitative reverse transcription polymerase chain reaction (qRT-PCR) assay was performed to detect the expression of miR-506 and Rho associated coiled-coil containing protein kinase 2 (ROCK2). Cell proliferation and apoptosis were evaluated by MTT assay and flow cytometry, respectively. Bioinformatics analysis and luciferase reporter assays were performed to identify the regulation between miR-506 and ROCK2. Western blot assay was performed to detect the expression of ROCK2, RhoA, and Ras-related C3 botulinum toxin substrate 1 (Rac1). The tumor growth in vivo was evaluated in a HCC xenograft mice model. RESULTS The mRNA levels of ROCK2 were significantly upregulated, while miR-506 levels were significantly downregulated in HCC tissues and cells. The expression of ROCK2 was negatively correlated with miR-506 in HCC tissues. In vitro, upregulation of miR-506 inhibited proliferation and induced apoptosis, and downregulation of miR-506 promoted proliferation and blocked apoptosis in HepG2 and Hep3B cells. ROCK2 was a target gene of miR-506 and miR-506 regulated the expression of ROCK2 in HepG2 and Hep3B cells. Furthermore, downregulation of miR-506 partially attenuated the tumor-suppressive effect of ROCK2 knockout on HepG2 and Hep3B cells, and upregulation of miR-506 partially attenuated the oncogenic effect of ROCK2 overexpression on HepG2 and Hep3B cells; Overexpression of ROCK2 increased and ROCK2 knockdown decreased the expression of Rac1, which were attenuated by upregulation of miR-506 or downregulation of miR-506, respectively. In addition, ROCK2 overexpression or knockdown hadno significant effect on RhoA expression. In vivo, upregulation of miR-506 suppressed tumor growth, while downregulation of miR-506 promoted tumor growth. CONCLUSION miR-506 was involved in cell proliferation and apoptosis by affecting RhoA/ROCK signaling pathway in HCC cells. Our results provide a novel mechanism of miR-506-mediated suppressive effects on HCC tumorigenesis.
Collapse
Affiliation(s)
- Linfei Zhang
- Department of Hepatobiliary Pancreatic Surgery, Renmin Hospital, Hubei University of MedicineShiyan 442000, Hubei, PR China
| | - Huadong Zhou
- Department of Hepatobiliary Pancreatic Surgery, Renmin Hospital, Hubei University of MedicineShiyan 442000, Hubei, PR China
| | - Gang Wei
- Department of Gastroenterology, Renmin Hospital, Hubei University of MedicineShiyan 442000, Hubei, PR China
| |
Collapse
|
190
|
Chronic administration of LIMK2 inhibitors alleviates cavernosal veno-occlusive dysfunction through suppression of cavernosal fibrosis in a rat model of erectile dysfunction after cavernosal nerve injury. PLoS One 2019; 14:e0213586. [PMID: 30870492 PMCID: PMC6417654 DOI: 10.1371/journal.pone.0213586] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 02/22/2019] [Indexed: 12/05/2022] Open
Abstract
We evaluated whether chronic administration of LIMK2-inhibitors could improve erectile function by alleviating CVOD through suppressing cavernosal fibrosis in a rat model of cavernosal nerve crush-injury (CNCI). Forty-two 12-week-old rats were equally categorized into the three groups: sham-surgery (S), CNCI (I), and CNCI treated with LIMK2-inhibitors (L). The L-group was treated with daily intraperitoneal injection of LIMK2-inhibitors (10.0 mg/kg) for 30-days after surgery. Erectile function was assessed using dynamic-infusion-cavernosometry (DIC). Penile tissue was processed for Masson’s-trichrome staining, Western-blotting, and double immunofluorescence. The I-group showed significantly higher maintenance and drop rates as well as lower papaverine response, compared to the S-group. Chronic inhibition of LIMK2 in the L-group significantly improved the DIC parameters compared to those in the I-group, although the parameters were not completely restored to normal control values. Also, the I-group showed a reduced smooth muscle (SM)-to-collagen ratio, decreased immunohistochemical staining for α-SM-actin, increased number of fibroblasts positive for phosphorylated Cofilin, increased LIMK2/Cofilin phosphorylation and increased protein expression of Collagen-1 or Fibronectin, compared to the S-group. The L-group showed significant improvements in SM/collagen ratio and the deposition of Collagen-1 or Fibronectin compared to the I-group, although not completely normalized. According to the densitometry and confocal microscopy results, the L-group showed restoration of LIMK2/Cofilin phosphorylation and amount of fibroblasts positive for phosphorylated Cofilin to the normal control value. In conclusion, chronic inhibition of LIMK2 can improve CVOD and ED by alleviating cavernosal fibrosis via normalizing the LIMK2/Cofilin pathway.
Collapse
|
191
|
Flentje A, Kalsi R, Monahan TS. Small GTPases and Their Role in Vascular Disease. Int J Mol Sci 2019; 20:ijms20040917. [PMID: 30791562 PMCID: PMC6413073 DOI: 10.3390/ijms20040917] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 01/31/2019] [Accepted: 02/07/2019] [Indexed: 12/18/2022] Open
Abstract
Over eighty million people in the United States have cardiovascular disease that can affect the heart causing myocardial infarction; the carotid arteries causing stroke; and the lower extremities leading to amputation. The treatment for end-stage cardiovascular disease is surgical—either endovascular therapy with balloons and stents—or open reconstruction to reestablish blood flow. All interventions damage or destroy the protective inner lining of the blood vessel—the endothelium. An intact endothelium is essential to provide a protective; antithrombotic lining of a blood vessel. Currently; there are no agents used in the clinical setting that promote reendothelialization. This process requires migration of endothelial cells to the denuded vessel; proliferation of endothelial cells on the denuded vessel surface; and the reconstitution of the tight adherence junctions responsible for the formation of an impermeable surface. These processes are all regulated in part and are dependent on small GTPases. As important as the small GTPases are for reendothelialization, dysregulation of these molecules can result in various vascular pathologies including aneurysm formation, atherosclerosis, diabetes, angiogenesis, and hypertension. A better understanding of the role of small GTPases in endothelial cell migration is essential to the development for novel agents to treat vascular disease.
Collapse
Affiliation(s)
- Alison Flentje
- Division of Vascular Surgery, Department of Surgery, University of Maryland School of Medicine, 22 South Greene Street, Suite S10B00, Baltimore, MD 21201, USA.
| | - Richa Kalsi
- Division of Vascular Surgery, Department of Surgery, University of Maryland School of Medicine, 22 South Greene Street, Suite S10B00, Baltimore, MD 21201, USA.
| | - Thomas S Monahan
- Division of Vascular Surgery, Department of Surgery, University of Maryland School of Medicine, 22 South Greene Street, Suite S10B00, Baltimore, MD 21201, USA.
| |
Collapse
|
192
|
Cantoni S, Cavalli S, Pastore F, Accetta A, Pala D, Vaccaro F, Cesari N, De Logu F, Nassini R, Villetti G, Facchinetti F. Pharmacological characterization of a highly selective Rho kinase (ROCK) inhibitor and its therapeutic effects in experimental pulmonary hypertension. Eur J Pharmacol 2019; 850:126-134. [PMID: 30753868 DOI: 10.1016/j.ejphar.2019.02.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/06/2019] [Accepted: 02/08/2019] [Indexed: 02/02/2023]
Abstract
Studies on the role of Rho-associated protein kinase (ROCK) in experimental pulmonary artery hypertension (PAH) relies mainly on the use of pharmacological inhibitors. However, interpreting these data is hampered by the lack of specificity of commonly utilized inhibitors. To fill this gap, we have selected and characterized a novel ROCK inhibitor, Compound 3, previously described in a patent. Inhibitory potency of Compound 3 against enzymatic activity of ROCK-1 and 2 (IC50 = 10 ± 3.1 and 7.8 ± 0.5 nM, respectively) was accompanied by a strong vasodilating effect in phenylephrine pre-contracted isolated rat pulmonary artery rings (IC50 = 51.7 ± 9.1 nM) as well as in aortic rings (IC50 = 45.5 ± 1.1 nM). Compound 3 showed a remarkable selectivity towards ROCK 1 and 2 when tested against a large panel (>400) of human kinases. A partial explanation for its selectivity is provided from docking simulations within ROCK-1. Pharmacokinetic studies showed that Compound 3 is suitable for a twice daily administration without significant accumulation upon repeated dosing. In rats with monocrotaline (MCT)-induced pulmonary hypertension, therapy with Compound 3, (1 and 3 mg/kg, s.c., b.i.d.), started 14 days after induction of the disease, attenuated right ventricle systolic pressure (RVSP) increase. Morphometric histological analysis showed that Compound 3, at both doses, counteracted MCT-induced medial thickening of lung distal arterioles with an effect comparable to macitentan (10 mg/kg, p.o., q.d.). Compound 3 is a potent and highly selective ROCK inhibitor that ameliorates hemodynamic parameters and counteracts pulmonary vascular remodeling in experimental PAH.
Collapse
Affiliation(s)
- Silvia Cantoni
- Corporate Pre-clinical R&D, Chiesi Farmaceutici S.p.A., Parma, Italy
| | - Stefano Cavalli
- Corporate Pre-clinical R&D, Chiesi Farmaceutici S.p.A., Parma, Italy
| | - Fiorella Pastore
- Corporate Pre-clinical R&D, Chiesi Farmaceutici S.p.A., Parma, Italy
| | - Alessandro Accetta
- Corporate Pre-clinical R&D, Chiesi Farmaceutici S.p.A., Parma, Italy
| | - Daniele Pala
- Corporate Pre-clinical R&D, Chiesi Farmaceutici S.p.A., Parma, Italy
| | - Fabio Vaccaro
- Corporate Pre-clinical R&D, Chiesi Farmaceutici S.p.A., Parma, Italy
| | - Nicola Cesari
- Corporate Pre-clinical R&D, Chiesi Farmaceutici S.p.A., Parma, Italy
| | - Francesco De Logu
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Romina Nassini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Gino Villetti
- Corporate Pre-clinical R&D, Chiesi Farmaceutici S.p.A., Parma, Italy
| | | |
Collapse
|
193
|
Wilmanns JC, Pandey R, Hon O, Chandran A, Schilling JM, Forte E, Wu Q, Cagnone G, Bais P, Philip V, Coleman D, Kocalis H, Archer SK, Pearson JT, Ramialison M, Heineke J, Patel HH, Rosenthal NA, Furtado MB, Costa MW. Metformin intervention prevents cardiac dysfunction in a murine model of adult congenital heart disease. Mol Metab 2019; 20:102-114. [PMID: 30482476 PMCID: PMC6358551 DOI: 10.1016/j.molmet.2018.11.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 11/06/2018] [Accepted: 11/10/2018] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE Congenital heart disease (CHD) is the most frequent birth defect worldwide. The number of adult patients with CHD, now referred to as ACHD, is increasing with improved surgical and treatment interventions. However the mechanisms whereby ACHD predisposes patients to heart dysfunction are still unclear. ACHD is strongly associated with metabolic syndrome, but how ACHD interacts with poor modern lifestyle choices and other comorbidities, such as hypertension, obesity, and diabetes, is mostly unknown. METHODS We used a newly characterized mouse genetic model of ACHD to investigate the consequences and the mechanisms associated with combined obesity and ACHD predisposition. Metformin intervention was used to further evaluate potential therapeutic amelioration of cardiac dysfunction in this model. RESULTS ACHD mice placed under metabolic stress (high fat diet) displayed decreased left ventricular ejection fraction. Comprehensive physiological, biochemical, and molecular analysis showed that ACHD hearts exhibited early changes in energy metabolism with increased glucose dependence as main cardiac energy source. These changes preceded cardiac dysfunction mediated by exposure to high fat diet and were associated with increased disease severity. Restoration of metabolic balance by metformin administration prevented the development of heart dysfunction in ACHD predisposed mice. CONCLUSIONS This study reveals that early metabolic impairment reinforces heart dysfunction in ACHD predisposed individuals and diet or pharmacological interventions can be used to modulate heart function and attenuate heart failure. Our study suggests that interactions between genetic and metabolic disturbances ultimately lead to the clinical presentation of heart failure in patients with ACHD. Early manipulation of energy metabolism may be an important avenue for intervention in ACHD patients to prevent or delay onset of heart failure and secondary comorbidities. These interactions raise the prospect for a translational reassessment of ACHD presentation in the clinic.
Collapse
Affiliation(s)
- Julia C Wilmanns
- Australian Regenerative Medicine Institute, Monash University, Australia; Department of Cardiology and Angiology, Experimental Cardiology, Hannover Medical School, Germany
| | | | | | - Anjana Chandran
- Australian Regenerative Medicine Institute, Monash University, Australia
| | - Jan M Schilling
- VA San Diego Healthcare System and Department of Anesthesiology, University of California San Diego, USA
| | | | - Qizhu Wu
- Monash Biomedical Imaging, Monash University, Australia
| | - Gael Cagnone
- Department of Pharmacology, Research Center of CHU Sainte-Justine, Canada
| | | | | | | | | | - Stuart K Archer
- Monash Bioinformatics Platform, Monash University, Australia; Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Australia
| | - James T Pearson
- Monash Biomedical Imaging, Monash University, Australia; Department of Physiology, Monash University, Australia; National Cerebral & Cardiovascular Center, Suita 565-8565, Japan
| | - Mirana Ramialison
- Australian Regenerative Medicine Institute, Monash University, Australia; Systems Biology Institute, Australia
| | - Joerg Heineke
- Department of Cardiology and Angiology, Experimental Cardiology, Hannover Medical School, Germany
| | - Hemal H Patel
- VA San Diego Healthcare System and Department of Anesthesiology, University of California San Diego, USA
| | - Nadia A Rosenthal
- The Jackson Laboratory, USA; Australian Regenerative Medicine Institute, Monash University, Australia; National Heart and Lung Institute, Imperial College London, W12 0NN, UK
| | - Milena B Furtado
- The Jackson Laboratory, USA; Australian Regenerative Medicine Institute, Monash University, Australia
| | - Mauro W Costa
- The Jackson Laboratory, USA; Australian Regenerative Medicine Institute, Monash University, Australia.
| |
Collapse
|
194
|
Werner JH, Rosenberg JH, Um JY, Moulton MJ, Agrawal DK. Molecular discoveries and treatment strategies by direct reprogramming in cardiac regeneration. Transl Res 2019; 203:73-87. [PMID: 30142308 PMCID: PMC6289806 DOI: 10.1016/j.trsl.2018.07.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 06/27/2018] [Accepted: 07/25/2018] [Indexed: 12/14/2022]
Abstract
Cardiac tissue has minimal endogenous regenerative capacity in response to injury. Treatment options are limited following tissue damage after events such as myocardial infarction. Current strategies are aimed primarily at injury prevention, but attention has been increasingly targeted toward the development of regenerative therapies. This review focuses on recent developments in the field of cardiac fibroblast reprogramming into induced cardiomyocytes. Early efforts to produce cardiac regeneration centered around induced pluripotent stem cells, but clinical translation has proved elusive. Currently, techniques are being developed to directly transdifferentiate cardiac fibroblasts into induced cardiomyocytes. Viral vector-driven expression of a combination of transcription factors including Gata4, Mef2c, and Tbx5 induced cardiomyocyte development in mice. Subsequent combinational modifications have extended these results to human cell lines and increased efficacy. The miRNAs including combinations of miR-1, miR-133, miR-208, and miR-499 can improve or independently drive regeneration of cardiomyocytes. Similar results could be obtained by combinations of small molecules with or without transcription factor or miRNA expression. The local tissue environment greatly impacts favorability for reprogramming. Modulation of signaling pathways, especially those mediated by VEGF and TGF-β, enhance differentiation to cardiomyocytes. Current reprogramming strategies are not ready for clinical application, but recent breakthroughs promise regenerative cardiac therapies in the near future.
Collapse
Affiliation(s)
- John H Werner
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska
| | - John H Rosenberg
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska
| | - John Y Um
- Department of Cardiothoracic Surgery, University of Nebraska Medical Center, Omaha, Nebraska
| | - Michael J Moulton
- Department of Cardiothoracic Surgery, University of Nebraska Medical Center, Omaha, Nebraska
| | - Devendra K Agrawal
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska.
| |
Collapse
|
195
|
Yin J, Lv L, Zhai P, Long T, Zhou Q, Pan H, Botwe G, Wang L, Wang Q, Tan L, Kuebler WM. Connexin 40 regulates lung endothelial permeability in acute lung injury via the ROCK1-MYPT1- MLC20 pathway. Am J Physiol Lung Cell Mol Physiol 2019; 316:L35-L44. [PMID: 30234377 DOI: 10.1152/ajplung.00012.2018] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Increased pulmonary vascular permeability is a hallmark of acute lung injury (ALI). Connexin 40 (Cx40) is a gap junctional protein abundantly present in the lung microvascular endothelium. Yet, the role of Cx40 in the regulation of lung vascular permeability and its underlying mechanisms are unclear. Here, we tested the hypothesis that Cx40 participates in regulation of lung endothelial permeability via a mechanism involving a Rho-associated protein kinase (ROCK) dependent regulation of myosin light chain (MLC). In murine models of intratracheal acid- or LPS-induced lung injury, genetic deficiency of Cx40 attenuated key features of ALI including vascular barrier failure. In human pulmonary microvascular endothelial cells (PMVECs), thrombin-induced loss of transendothelial electrical resistance was attenuated by a Cx40-inhibiting mimetic peptide (40GAP27), Cx40-specific shRNA, or ROCK inhibitor Y27632. In isolated perfused mouse lungs, platelet-activating factor-induced lung weight gain was abrogated by gap junction blocker carbenoxolone, 40GAP27, Y27632, or genetic deficiency of Cx40. Phosphorylation of MLC20 increased drastically in both LPS-treated PMVECs and HCl-treated mouse lungs. Expression of ROCK1 was increased in both LPS-treated PMVECs and HCl-treated mouse lungs, and paralleled by phosphorylation of MLC20. Coimmunoprecipitation experiments revealed protein-protein interaction between ROCK1 and Cx40. LPS-induced upregulation of ROCK1 and phosphorylation of MLC20 were blocked by knockdown of Cx40. LPS caused phosphorylation of myosin phosphatase targeting subunit 1, which could be abrogated by Y27632 or Cx40-shRNA. Our findings reveal a role of Cx40 in regulation of ROCK1 and MLC20 that contributes critically to lung vascular barrier failure in ALI.
Collapse
Affiliation(s)
- Jun Yin
- Department of Thoracic Surgery, Zhongshan Hospital of Fudan University , Shanghai , China
- Department of Cardiothoracic Surgery, Affiliated People's Hospital of Jiangsu University , Zhenjiang, Jiangsu , China
| | - Lu Lv
- Department of Cardiothoracic Surgery, Affiliated People's Hospital of Jiangsu University , Zhenjiang, Jiangsu , China
| | - Peng Zhai
- Department of Cardiothoracic Surgery, Affiliated People's Hospital of Jiangsu University , Zhenjiang, Jiangsu , China
| | - Tao Long
- Department of Cardiothoracic Surgery, Affiliated People's Hospital of Jiangsu University , Zhenjiang, Jiangsu , China
| | - Qiang Zhou
- Department of Cardiothoracic Surgery, Affiliated People's Hospital of Jiangsu University , Zhenjiang, Jiangsu , China
| | - Huiwen Pan
- Department of Cardiothoracic Surgery, Affiliated People's Hospital of Jiangsu University , Zhenjiang, Jiangsu , China
| | - Godwin Botwe
- Department of Cardiothoracic Surgery, Affiliated People's Hospital of Jiangsu University , Zhenjiang, Jiangsu , China
| | - Liming Wang
- Department of Chemotherapy, Cancer Institute, Affiliated People's Hospital of Jiangsu University , Zhenjiang, Jiangsu , China
| | - Qun Wang
- Department of Thoracic Surgery, Zhongshan Hospital of Fudan University , Shanghai , China
| | - Lijie Tan
- Department of Thoracic Surgery, Zhongshan Hospital of Fudan University , Shanghai , China
| | - Wolfgang M Kuebler
- Department of Physiology and Center for Cardiovascular Research, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health , Berlin , Germany
| |
Collapse
|
196
|
Afewerki T, Ahmed S, Warren D. Emerging regulators of vascular smooth muscle cell migration. J Muscle Res Cell Motil 2019; 40:185-196. [PMID: 31254136 PMCID: PMC6726670 DOI: 10.1007/s10974-019-09531-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 06/21/2019] [Indexed: 12/30/2022]
Abstract
Vascular smooth muscle cells (VSMCs) are the predominant cell type in the blood vessel wall and normally adopt a quiescent, contractile phenotype. VSMC migration is tightly controlled, however, disease associated changes in the soluble and insoluble environment promote VSMC migration. Classically, studies investigating VSMC migration have described the influence of soluble factors. Emerging data has highlighted the importance of insoluble factors, including extracellular matrix stiffness and porosity. In this review, we will recap on the important signalling pathways that regulate VSMC migration and reflect on the potential importance of emerging regulators of VSMC function.
Collapse
Affiliation(s)
- TecLino Afewerki
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ UK
| | - Sultan Ahmed
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ UK
| | - Derek Warren
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ UK
| |
Collapse
|
197
|
de Oliveira RG, Guerra FS, Mermelstein CDS, Fernandes PD, Bastos ITDS, Costa FN, Barroso RCR, Ferreira FF, Fraga CAM. Synthesis and pharmacological evaluation of novel isoquinoline N-sulphonylhydrazones designed as ROCK inhibitors. J Enzyme Inhib Med Chem 2018; 33:1181-1193. [PMID: 30044647 PMCID: PMC6060383 DOI: 10.1080/14756366.2018.1490732] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 06/14/2018] [Accepted: 06/15/2018] [Indexed: 12/31/2022] Open
Abstract
In this study, we synthesized a new congener series of N-sulphonylhydrazones designed as candidate ROCK inhibitors using the molecular hybridization of the clinically approved drug fasudil (1) and the IKK-β inhibitor LASSBio-1524 (2). Among the synthesized compounds, the N-methylated derivative 11 (LASSBio-2065) showed the best inhibitory profile for both ROCK isoforms, with IC50 values of 3.1 and 3.8 µM for ROCK1 and ROCK2, respectively. Moreover, these compounds were also active in the scratch assay performed in human breast cancer MDA-MB 231 cells and did not display toxicity in MTT and LDH assays. Molecular modelling studies provided insights into the possible binding modes of these N-sulphonylhydrazones, which present a new molecular architecture capable of being optimized and developed as therapeutically useful ROCK inhibitors.
Collapse
Affiliation(s)
- Ramon Guerra de Oliveira
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fabiana Sélos Guerra
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratório de Farmacologia da Dor e da Inflamação, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cláudia dos Santos Mermelstein
- Laboratório de Diferenciação Muscular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patrícia Dias Fernandes
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratório de Farmacologia da Dor e da Inflamação, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Fanny Nascimento Costa
- Centro de Ciências Naturais e Humanas (CCNH), Universidade Federal do ABC (UFABC), São Paulo, Brazil
| | | | - Fabio Furlan Ferreira
- Centro de Ciências Naturais e Humanas (CCNH), Universidade Federal do ABC (UFABC), São Paulo, Brazil
| | - Carlos Alberto Manssour Fraga
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
198
|
Sonic hedgehog regulation of cavernous nerve regeneration and neurite formation in aged pelvic plexus. Exp Neurol 2018; 312:10-19. [PMID: 30391523 DOI: 10.1016/j.expneurol.2018.11.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 09/27/2018] [Accepted: 11/01/2018] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Erectile dysfunction (ED) is a significant health concern that greatly impacts quality of life, and is common in men as they age, impacting 52% of men between the ages of 40 and 70. A significant underlying cause of ED development is injury to the cavernous nerve (CN), a peripheral nerve that innervates the penis. CN injury also occurs in up to 82% of prostatectomy patients. We recently showed that Sonic hedgehog (SHH) protein delivered by peptide amphiphile (PA) nanofiber hydrogel to the CN and penis of a prostatectomy model of CN injury, is neuroprotective, accelerates CN regeneration, improves erectile function ~60%, preserves penile smooth muscle 56% and suppresses collagen deposition 30%. This regenerative potential is substantial in an adult prostatectomy model (P120). However prostatectomy patients are typically older (61.5 ± 9.6 years) and our models should mimic patient conditions more effectively when considering translation. In this study we examine regenerative potential in an aged prostatectomy model (P200-329). METHODS The caudal portion of the pelvic ganglia (MPG) and CN were dissected from adult (n = 11), and aged (n = 13) Sprague Dawley rats, and were grown in organ culture 3 days. Uninjured and 2 day CN crushed MPG/CN were exposed to Affi-Gel beads containing SHH protein, PBS (control), or 5e1 SHH inhibitor. Neurites were quantified by counting the number of growth cones normalized by tissue perimeter (mm) and immunohistochemistry for SHH, patched1 (PTCH1), smoothened (SMO), GLI1-3, and GAP43 were performed. RESULTS SHH treatment increased neurites 3.5-fold, in uninjured adult, and 5.7-fold in aged rats. Two days after CN crush, SHH treatment increased neurites 1.8-fold in adult rats and 2.5-fold in aged rats. SHH inhibition inhibited neurite formation in uninjured MPG/CN but not in 2 day CN crushed MPG/CN. PTCH1 and SMO (SHH receptors), and SHH transcriptional activators/repressors, GLI1-3, were abundant in aged MPG/CN with unaltered localization. ROCK1 was induced with SHH treatment. CONCLUSIONS Reintroduction of SHH protein in an aged prostatectomy model is even more effective in promoting neurite formation/CN regeneration than in the adult. The first 48 h after CN injury are a critical window when growth factors are released, that impact later neurite formation. These studies are significant because most prostatectomy patients are not young and healthy, as with adult rats, so the aged prostatectomy model will more accurately simulate ED patient response. Understanding how neurite formation changes with age is critical for clinical translation of SHH PA to prostatectomy patients.
Collapse
|
199
|
Ashwinbalaji S, Senthilkumari S, Gowripriya C, Krishnadas S, Gabelt BAT, Kaufman PL, Muthukkaruppan V. SB772077B, A New Rho Kinase Inhibitor Enhances Aqueous Humour Outflow Facility in Human Eyes. Sci Rep 2018; 8:15472. [PMID: 30341380 PMCID: PMC6195566 DOI: 10.1038/s41598-018-33932-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 10/08/2018] [Indexed: 11/09/2022] Open
Abstract
We investigated the effect of a new Rho kinase inhibitor, SB772077B (SB77) on aqueous outflow facility (OF) in human eyes using human organ-cultured anterior segment (HOCAS). IOP was monitored for 24 h post-treatment with either SB77 (0.1/10/50 µM) or vehicle after a stable baseline pressure. The hydrodynamic pattern of aqueous outflow was analysed by labelling outflow pathway with red fluorescent microspheres. The effect of SB77 on cell morphology, actin stress fibers, focal adhesions, ECM, status of RhoA activation and myosin light chain phosphorylation (p-MLC) were evaluated and compared with Y27632, by immunostaining using primary human trabecular meshwork (HTM) cells. Following 24 h treatment, SB77 increased OF by 16% at 0.1 µM (N = 6), 29% at 10 µM (N = 8; p = 0.018) and 39% at 50 µM (N = 8; p = 0.004) in human eyes. There was an overall increase in tracer quantity and in area along inner wall of Schlemm’s canal. Treatment with SB77 showed no evidence of cytotoxicity and caused a significant reduction in the expression of fibrotic markers compared to Y27632. The present findings indicate that SB77 treatment was effective in enhancing OF and reducing fibrotic markers in an ex vivo model. Thus SB77 may be a potential clinical candidate for the management of glaucoma.
Collapse
Affiliation(s)
- Soundararajan Ashwinbalaji
- Department of Ocular Pharmacology, Aravind Medical Research Foundation, #1, Anna Nagar, Madurai-20, Tamilnadu, India
| | - Srinivasan Senthilkumari
- Department of Ocular Pharmacology, Aravind Medical Research Foundation, #1, Anna Nagar, Madurai-20, Tamilnadu, India.
| | - Chidambaranathan Gowripriya
- Department of Immunology & Stem Cell Biology, Aravind Medical Research Foundation, #1, Anna Nagar, Madurai-20, Tamilnadu, India
| | - Subbaiah Krishnadas
- Glaucoma Clinic, Aravind Eye Hospital, #1, Anna Nagar, Madurai-20, Tamilnadu, India
| | - B' Ann T Gabelt
- Department of Ophthalmology & Visual Sciences, University of Wisconsin, Madison, Wisconsin, USA
| | - Paul L Kaufman
- Department of Ophthalmology & Visual Sciences, University of Wisconsin, Madison, Wisconsin, USA
| | | |
Collapse
|
200
|
Rho kinase activation in circulating leukocytes is related to hypertensive myocardial remodeling. Clin Sci (Lond) 2018; 132:1837-1853. [PMID: 30065083 DOI: 10.1042/cs20180312] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 07/19/2018] [Accepted: 07/23/2018] [Indexed: 12/18/2022]
Abstract
Rho-kinase has relevant functions in blood pressure modulation and cardiovascular remodeling. Rho-kinase activity is determined in circulating leukocytes measuring phosphorylation of its target myosin phosphatase target subunit 1 (MYPT1), but its relationship with Rho-kinase activity in the myocardium and in vasculature in hypertension has not been evaluated.The aim was to determine the degree of association between Rho-kinase cascade activation in circulating leukocytes with cardiac and aortic Rho-kinase pathway activation in a model of hypertension and to analyze it with a cause-effect perspective.Hypertensive deoxycorticosterone (DOCA)-salt rats received the Rho-kinase antagonist fasudil (DOCA-Fas, 100 mg/kg/day, 3 weeks). Results were compared with an untreated DOCA-salt and a sham group.Rho-kinase inhibition reduced significantly blood pressure, cardiac hypertrophy, myocardial collagen and macrophage infiltration, but not aortic wall hypertrophy. Fasudil decreased significantly Rho-kinase activity in peripheral blood mononucleated cells (PBMC), myocardium and aortic wall to similar levels as in the sham group. A significant correlation was found between PBMC Rho-kinase activity and cardiac remodeling, specifically with hypertrophy (r = 0.51, P≤0.01), myocardial collagen (r = 0.40, P≤0.05) and ED1 immunostaining (r = 0.48, P≤0.01). In the untreated hypertensive group, increased levels (P<0.05) of the proinflammatory molecules p65 NF-κB, vascular cell adhesion molecule 1 and interleukin-6 antibody in the myocardium, aortic wall and PBMC were observed and were reduced with fasudil (P<0.05).In conclusion, in this hypertension model, Rho-kinase and its pathway activation determined in circulating leukocytes reflect the activation of this pathway in the myocardium and in the aortic wall and are significantly related to myocardial remodeling (hypertrophy, fibrosis and inflammation).
Collapse
|