1
|
Blazanin N, Liang X, Mahmud I, Kim E, Martinez S, Tan L, Chan W, Anvar NE, Ha MJ, Qudratullah M, Minelli R, Peoples M, Lorenzi P, Hart T, Lissanu Y. Therapeutic modulation of ROCK overcomes metabolic adaptation of cancer cells to OXPHOS inhibition and drives synergistic anti-tumor activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.16.613317. [PMID: 39345502 PMCID: PMC11429714 DOI: 10.1101/2024.09.16.613317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Genomic studies have identified frequent mutations in subunits of the SWI/SNF chromatin remodeling complex including SMARCA4 and ARID1A in non-small cell lung cancer. Previously, we and others have identified that SMARCA4-mutant lung cancers are highly dependent on oxidative phosphorylation (OXPHOS). Despite initial excitements, therapeutics targeting metabolic pathways such as OXPHOS have largely been disappointing due to rapid adaptation of cancer cells to inhibition of single metabolic enzymes or pathways, suggesting novel combination strategies to overcome adaptive responses are urgently needed. Here, we performed a functional genomics screen using CRISPR-Cas9 library targeting genes with available FDA approved therapeutics and identified ROCK1/2 as a top hit that sensitizes cancer cells to OXPHOS inhibition. We validate these results by orthogonal genetic and pharmacologic approaches by demonstrating that KD025 (Belumosudil), an FDA approved ROCK inhibitor, has highly synergistic anti-cancer activity in vitro and in vivo in combination with OXPHOS inhibition. Mechanistically, we showed that this combination induced a rapid, profound energetic stress and cell cycle arrest that was in part due to ROCK inhibition-mediated suppression of the adaptive increase in glycolysis normally seen by OXPHOS inhibition. Furthermore, we applied global phosphoproteomics and kinase-motif enrichment analysis to uncover a dynamic regulatory kinome upon combination of OXPHOS and ROCK inhibition. Importantly, we found converging phosphorylation-dependent regulatory cross-talk by AMPK and ROCK kinases on key RHO GTPase signaling/ROCK-dependent substrates such as PPP1R12A, NUMA1 and PKMYT1 that are known regulators of cell cycle progression. Taken together, our study identified ROCK kinases as critical mediators of metabolic adaptation of cancer cells to OXPHOS inhibition and provides a strong rationale for pursuing ROCK inhibitors as novel combination partners to OXPHOS inhibitors in cancer treatment.
Collapse
Affiliation(s)
- Nicholas Blazanin
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center
| | - Xiaobing Liang
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center
| | - Iqbal Mahmud
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center
| | - Eiru Kim
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center
| | - Sara Martinez
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center
| | - Lin Tan
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center
| | - Waikin Chan
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center
| | - Nazanin Esmaeili Anvar
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center
| | - Min Jin Ha
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Md Qudratullah
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center
| | - Rosalba Minelli
- TRACTION Platform, Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Michael Peoples
- TRACTION Platform, Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Philip Lorenzi
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center
| | - Traver Hart
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center
| | - Yonathan Lissanu
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center
| |
Collapse
|
2
|
Shi J, Wei L. ROCK1 deficiency preserves caveolar compartmentalization of signaling molecules and cell membrane integrity. FASEB Bioadv 2024; 6:85-102. [PMID: 38463696 PMCID: PMC10918988 DOI: 10.1096/fba.2024-00015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/05/2024] [Accepted: 02/08/2024] [Indexed: 03/12/2024] Open
Abstract
In this study, we investigated the roles of ROCK1 in regulating structural and functional features of caveolae located at the cell membrane of cardiomyocytes, adipocytes, and mouse embryonic fibroblasts (MEFs) as well as related physiopathological effects. Caveolae are small bulb-shaped cell membrane invaginations, and their roles have been associated with disease conditions. One of the unique features of caveolae is that they are physically linked to the actin cytoskeleton that is well known to be regulated by RhoA/ROCKs pathway. In cardiomyocytes, we observed that ROCK1 deficiency is coincident with an increased caveolar density, clusters, and caveolar proteins including caveolin-1 and -3. In the mouse cardiomyopathy model with transgenic overexpressing Gαq in myocardium, we demonstrated the reduced caveolar density at cell membrane and reduced caveolar protein contents. Interestingly, coexisting ROCK1 deficiency in cardiomyocytes can rescue these defects and preserve caveolar compartmentalization of β-adrenergic signaling molecules including β1-adrenergic receptor and type V/VI adenylyl cyclase. In cardiomyocytes and adipocytes, we detected that ROCK1 deficiency increased insulin signaling with increased insulin receptor activation in caveolae. In MEFs, we identified that ROCK1 deficiency increased caveolar and total levels of caveolin-1 and cell membrane repair ability after mechanical or chemical disruptions. Together, these results demonstrate that ROCK1 can regulate caveolae plasticity and multiple functions including compartmentalization of signaling molecules and cell membrane repair following membrane disruption by mechanical force and oxidative damage. These findings provide possible molecular insights into the beneficial effects of ROCK1 deletion/inhibition in cardiomyocytes, adipocytes, and MEFs under certain diseased conditions.
Collapse
Affiliation(s)
- Jianjian Shi
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, School of MedicineIndiana UniversityIndianapolisIndianaUSA
| | - Lei Wei
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, School of MedicineIndiana UniversityIndianapolisIndianaUSA
| |
Collapse
|
3
|
Shen J, Wang Y, Deng X, Sana SRGL. Combining bioinformatics and machine learning algorithms to identify and analyze shared biomarkers and pathways in COVID-19 convalescence and diabetes mellitus. Front Endocrinol (Lausanne) 2023; 14:1306325. [PMID: 38169604 PMCID: PMC10758397 DOI: 10.3389/fendo.2023.1306325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 12/01/2023] [Indexed: 01/05/2024] Open
Abstract
Background Most patients who had coronavirus disease 2019 (COVID-19) fully recovered, but many others experienced acute sequelae or persistent symptoms. It is possible that acute COVID-19 recovery is just the beginning of a chronic condition. Even after COVID-19 recovery, it may lead to the exacerbation of hyperglycemia process or a new onset of diabetes mellitus (DM). In this study, we used a combination of bioinformatics and machine learning algorithms to investigate shared pathways and biomarkers in DM and COVID-19 convalescence. Methods Gene transcriptome datasets of COVID-19 convalescence and diabetes mellitus from Gene Expression Omnibus (GEO) were integrated using bioinformatics methods and differentially expressed genes (DEGs) were found using the R programme. These genes were also subjected to Gene Ontology (GO) functional enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis to find potential pathways. The hub DEGs genes were then identified by combining protein-protein interaction (PPI) networks and machine learning algorithms. And transcription factors (TFs) and miRNAs were predicted for DM after COVID-19 convalescence. In addition, the inflammatory and immune status of diabetes after COVID-19 convalescence was assessed by single-sample gene set enrichment analysis (ssGSEA). Results In this study, we developed genetic diagnostic models for 6 core DEGs beteen type 1 DM (T1DM) and COVID-19 convalescence and 2 core DEGs between type 2 DM (T2DM) and COVID-19 convalescence and demonstrated statistically significant differences (p<0.05) and diagnostic validity in the validation set. Analysis of immune cell infiltration suggests that a variety of immune cells may be involved in the development of DM after COVID-19 convalescence. Conclusion We identified a genetic diagnostic model for COVID-19 convalescence and DM containing 8 core DEGs and constructed a nomogram for the diagnosis of COVID-19 convalescence DM.
Collapse
Affiliation(s)
- Jinru Shen
- The First Clinical Medical School, Harbin Medical University, Harbin, China
| | - Yaolou Wang
- The First Clinical Medical School, Harbin Medical University, Harbin, China
| | - Xijin Deng
- Department of Anaesthesiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Si Ri Gu Leng Sana
- Department of Anaesthesiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
4
|
Zanin-Zhorov A, Chen W, Moretti J, Nyuydzefe MS, Zhorov I, Munshi R, Ghosh M, Serdjebi C, MacDonald K, Blazar BR, Palmer M, Waksal SD. Selectivity matters: selective ROCK2 inhibitor ameliorates established liver fibrosis via targeting inflammation, fibrosis, and metabolism. Commun Biol 2023; 6:1176. [PMID: 37980369 PMCID: PMC10657369 DOI: 10.1038/s42003-023-05552-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 11/07/2023] [Indexed: 11/20/2023] Open
Abstract
The pathogenesis of hepatic fibrosis is driven by dysregulated metabolism precipitated by chronic inflammation. Rho-associated coiled-coil-containing protein kinases (ROCKs) have been implicated in these processes, however the ability of selective ROCK2 inhibition to target simultaneously profibrotic, pro-inflammatory and metabolic pathways remains undocumented. Here we show that therapeutic administration of GV101, a selective ROCK2 inhibitor with more than 1000-fold selectivity over ROCK1, attenuates established liver fibrosis induced by thioacetamide (TAA) in combination with high-fat diet in mice. GV101 treatment significantly reduces collagen levels in liver, associated with downregulation of pCofilin, pSTAT3, pAkt, while pSTAT5 and pAMPK levels are increased in tissues of treated mice. In vitro, GV101 inhibits profibrogenic markers expression in fibroblasts, adipogenesis in primary adipocytes and TLR-induced cytokine secretion in innate immune cells via targeting of Akt-mTOR-S6K signaling axis, further uncovering the ROCK2-specific complex mechanism of action and therapeutic potential of highly selective ROCK2 inhibitors in liver fibrosis.
Collapse
Affiliation(s)
| | - Wei Chen
- Graviton Bioscience B.V, Amsterdam, 1017 CG, Netherlands
| | - Julien Moretti
- Graviton Bioscience B.V, Amsterdam, 1017 CG, Netherlands
| | | | - Iris Zhorov
- Graviton Bioscience B.V, Amsterdam, 1017 CG, Netherlands
| | | | | | | | - Kelli MacDonald
- QIMR Berghofer Medical Research Institute, Brisbane, 4006, Australia
| | - Bruce R Blazar
- Division of Blood & Marrow Transplant & Cellular Therapies, University of MN, Masonic Cancer Center and Department of Pediatrics, Minneapolis, MN, 55455, USA
| | | | | |
Collapse
|
5
|
Dohnalkova E, Bayer RL, Guo Q, Bamidele AO, Kim Lee HS, Valenzuela-Pérez L, Krishnan A, Pavelko KD, Guisot NES, Bunyard P, Kim YB, Ibrahim SH, Gores GJ, Hirsova P. Rho-associated protein kinase 1 inhibition in hepatocytes attenuates nonalcoholic steatohepatitis. Hepatol Commun 2023; 7:02009842-202306010-00031. [PMID: 37267252 DOI: 10.1097/hc9.0000000000000171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/28/2023] [Indexed: 06/04/2023] Open
Abstract
BACKGROUND NASH is the progressive form of NAFLD characterized by lipotoxicity, hepatocyte injury, tissue inflammation, and fibrosis. Previously, Rho-associated protein kinase (ROCK) 1 has been implicated in lipotoxic signaling in hepatocytes in vitro and high-fat diet-induced lipogenesis in vivo. However, whether ROCK1 plays a role in liver inflammation and fibrosis during NASH is unclear. Here, we hypothesized that pathogenic activation of ROCK1 promotes murine NASH pathogenesis. METHODS AND RESULTS Patients with NASH had increased hepatic ROCK1 expression compared with patients with fatty liver. Similarly, hepatic ROCK1 levels and activity were increased in mice with NASH induced by a western-like diet that is high in fat, fructose, and cholesterol (FFC). Hepatocyte-specific ROCK1 knockout mice on the FFC diet displayed a decrease in liver steatosis, hepatic cell death, liver inflammation, and fibrosis compared with littermate FFC-fed controls. Mechanistically, these effects were associated with a significant attenuation of myeloid cell recruitment. Interestingly, myeloid cell-specific ROCK1 deletion did not affect NASH development in FFC-fed mice. To explore the therapeutic opportunities, mice with established NASH received ROCKi, a novel small molecule kinase inhibitor of ROCK1/2, which preferentially accumulates in liver tissue. ROCK inhibitor treatment ameliorated insulin resistance and decreased liver injury, inflammation, and fibrosis. CONCLUSIONS Genetic or pharmacologic inhibition of ROCK1 activity attenuates murine NASH, suggesting that ROCK1 may be a therapeutic target for treating human NASH.
Collapse
Affiliation(s)
- Ester Dohnalkova
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Biological and Medical Sciences, Charles University, Hradec Kralove, Czech Republic
| | - Rachel L Bayer
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Qianqian Guo
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Adebowale O Bamidele
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Hyun Se Kim Lee
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Anuradha Krishnan
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Kevin D Pavelko
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | - Young-Bum Kim
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Samar H Ibrahim
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
- Division of Pediatric Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Gregory J Gores
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Petra Hirsova
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
6
|
Sung BJ, Lim SB, Yang WM, Kim JH, Kulkarni RN, Kim YB, Lee MK. ROCK1 regulates insulin secretion from β-cells. Mol Metab 2022; 66:101625. [PMID: 36374631 PMCID: PMC9649378 DOI: 10.1016/j.molmet.2022.101625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/21/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVE The endocrine pancreatic β-cells play a pivotal role in maintaining whole-body glucose homeostasis and its dysregulation is a consistent feature in all forms of diabetes. However, knowledge of intracellular regulators that modulate β-cell function remains incomplete. We investigated the physiological role of ROCK1 in the regulation of insulin secretion and glucose homeostasis. METHODS Mice lacking ROCK1 in pancreatic β-cells (RIP-Cre; ROCK1loxP/loxP, β-ROCK1-/-) were studied. Glucose and insulin tolerance tests as well as glucose-stimulated insulin secretion (GSIS) were measured. An insulin secretion response to a direct glucose or pyruvate or pyruvate kinase (PK) activator stimulation in isolated islets from β-ROCK1-/- mice or β-cell lines with knockdown of ROCK1 was also evaluated. A proximity ligation assay was performed to determine the physical interactions between PK and ROCK1. RESULTS Mice with a deficiency of ROCK1 in pancreatic β-cells exhibited significantly increased blood glucose levels and reduced serum insulin without changes in body weight. Interestingly, β-ROCK1-/- mice displayed a progressive impairment of glucose tolerance while maintaining insulin sensitivity mostly due to impaired GSIS. Consistently, GSIS markedly decreased in ROCK1-deficient islets and ROCK1 knockdown INS-1 cells. Concurrently, ROCK1 blockade led to a significant decrease in intracellular calcium and ATP levels and oxygen consumption rates in isolated islets and INS-1 cells. Treatment of ROCK1-deficient islets or ROCK1 knockdown β-cells either with pyruvate or a PK activator rescued the impaired GSIS. Mechanistically, we observed that glucose stimulation in β-cells greatly enhanced ROCK1 binding to PK. CONCLUSIONS Our findings demonstrate that β-cell ROCK1 is essential for glucose-stimulated insulin secretion and for glucose homeostasis and that ROCK1 acts as an upstream regulator of glycolytic pyruvate kinase signaling.
Collapse
Affiliation(s)
- Byung-Jun Sung
- Division of Endocrinology and Metabolism, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.
| | - Sung-Bin Lim
- Division of Endocrinology and Metabolism, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.
| | - Won-Mo Yang
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.
| | - Jae Hyeon Kim
- Division of Endocrinology and Metabolism, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.
| | - Rohit N Kulkarni
- Islet Cell and Regenerative Medicine, Joslin Diabetes Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Stem Cell Institute, and Harvard Medical School, Boston, MA, USA.
| | - Young-Bum Kim
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.
| | - Moon-Kyu Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Nowon Eulji University Hospital, Eulji University School of Medicine, Seoul, South Korea.
| |
Collapse
|
7
|
Yang L, Qiao P, Zhang J, Huang S, Hu A. Rho-associated kinase1 promotes laryngeal squamous cell carcinoma tumorigenesis and progression via the FAK signaling pathway. Discov Oncol 2022; 13:100. [PMID: 36197602 PMCID: PMC9535064 DOI: 10.1007/s12672-022-00561-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/08/2022] [Indexed: 11/04/2022] Open
Abstract
Laryngeal squamous cell carcinoma (LSCC) is one of the most common head and neck squamous cell carcinomas (HNSCC). Rho-associated kinase1 (ROCK1) is considered to promote progression of numerous cancers, however, its role in LSCC is still unknown. Here, the expression level of ROCK1 is higher in LSCC tissues than non-tumor tissues, and the expression level of ROCK1 is positively correlated with advanced stage and poor survival prognosis. ROCK1 knockdown in TU686 and TU212 cells dramatically inhibits cellular proliferation, migration and invasion. Whereas the overexpression of ROCK1 reversed these changes. FAK signaling pathway plays an essential role in promoting LSCC progression. Inhibiting FAK activity with TAE226 observably impairs the tumor-promoting effects. In conclusion, ROCK1 promotes LSCC tumorigenesis and progression via the FAK signaling pathway, targeting the ROCK1 molecule may represent potential targets for clinical LSCC treatment.
Collapse
Affiliation(s)
- Liyun Yang
- Department of Otolaryngology, Gongli Hospital, The Second Military Medical University, Shanghai, 200135, China
| | - Peipei Qiao
- Department of Otolaryngology, Gongli Hospital, The Second Military Medical University, Shanghai, 200135, China
| | - Jianwei Zhang
- Department of Otolaryngology, Gongli Hospital, The Second Military Medical University, Shanghai, 200135, China
| | - Shuixian Huang
- Department of Otolaryngology, Gongli Hospital, The Second Military Medical University, Shanghai, 200135, China.
| | - An Hu
- Department of Otolaryngology, Gongli Hospital, The Second Military Medical University, Shanghai, 200135, China.
| |
Collapse
|
8
|
Penumatsa KC, Singhal AA, Warburton RR, Bear MD, Bhedi CD, Nasirova S, Wilson JL, Qi G, Preston IR, Hill NS, Fanburg BL, Kim YB, Toksoz D. Vascular smooth muscle ROCK1 contributes to hypoxia-induced pulmonary hypertension development in mice. Biochem Biophys Res Commun 2022; 604:137-143. [PMID: 35303680 PMCID: PMC9047112 DOI: 10.1016/j.bbrc.2022.02.064] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 02/18/2022] [Indexed: 11/26/2022]
Abstract
Rho kinase (ROCK) is implicated in the development of pulmonary arterial hypertension (PAH) in which abnormal pulmonary vascular smooth muscle (VSM) contractility and remodeling lead to right heart failure. Pharmacologic ROCK inhibitors block experimental pulmonary hypertension (PH) development in rodents but can have off-target effects and do not distinguish between the two ROCK forms, ROCK1 and ROCK2, encoded by separate genes. An earlier study using gene knock out (KO) in mice indicated that VSM ROCK2 is required for experimental PH development, but the role of ROCK1 is not well understood. Here we investigated the in vivo role of ROCK1 in PH development by generating a VSM-targeted homozygous ROCK1 gene KO mouse strain. Adult control mice exposed to Sugen5416 (Su)/hypoxia treatment to induce PH had significantly increased right ventricular systolic pressures (RVSP) and RV hypertrophy versus normoxic controls. In contrast, Su/hypoxia-exposed VSM ROCK1 KO mice did not exhibit significant RVSP elevation, and RV hypertrophy was blunted. Su/hypoxia-induced pulmonary small vessel muscularization was similarly elevated in both control and VSM ROCK1 KO animals. siRNA-mediated ROCK1 knock-down (KD) in human PAH pulmonary arterial SM cells (PASMC) did not affect cell growth. However, ROCK1 KD led to reduced AKT and MYPT1 signaling in serotonin-treated PAH PASMC. The findings suggest that like VSM ROCK2, VSM ROCK1 actively contributes to PH development, but in distinction acts via nonproliferative pathways to promote hypoxemia, and thus may be a distinct therapeutic target in PH.
Collapse
Affiliation(s)
- Krishna C Penumatsa
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA
| | - Adit A Singhal
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA
| | - Rod R Warburton
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA
| | - Michael D Bear
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA
| | - Chinmayee D Bhedi
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA
| | - Sabina Nasirova
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA
| | - Jamie L Wilson
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA
| | - Guanming Qi
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA
| | - Ioana R Preston
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA
| | - Nicholas S Hill
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA
| | - Barry L Fanburg
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA
| | - Young-Bum Kim
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, 330 Brookline Ave., Boston, MA, 02115, USA
| | - Deniz Toksoz
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA.
| |
Collapse
|
9
|
Wei L, Shi J. Insight Into Rho Kinase Isoforms in Obesity and Energy Homeostasis. Front Endocrinol (Lausanne) 2022; 13:886534. [PMID: 35769086 PMCID: PMC9234286 DOI: 10.3389/fendo.2022.886534] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/06/2022] [Indexed: 11/13/2022] Open
Abstract
Obesity and associated complications increasingly jeopardize global health and contribute to the rapidly rising prevalence of type 2 diabetes mellitus and obesity-related diseases. Developing novel methods for the prevention and treatment of excess body adipose tissue expansion can make a significant contribution to public health. Rho kinase is a Rho-associated coiled-coil-containing protein kinase (Rho kinase or ROCK). The ROCK family including ROCK1 and ROCK2 has recently emerged as a potential therapeutic target for the treatment of metabolic disorders. Up-regulated ROCK activity has been involved in the pathogenesis of all aspects of metabolic syndrome including obesity, insulin resistance, dyslipidemia and hypertension. The RhoA/ROCK-mediated actin cytoskeleton dynamics have been implicated in both white and beige adipogenesis. Studies using ROCK pan-inhibitors in animal models of obesity, diabetes, and associated complications have demonstrated beneficial outcomes. Studies via genetically modified animal models further established isoform-specific roles of ROCK in the pathogenesis of metabolic disorders including obesity. However, most reported studies have been focused on ROCK1 activity during the past decade. Due to the progress in developing ROCK2-selective inhibitors in recent years, a growing body of evidence indicates more attention should be devoted towards understanding ROCK2 isoform function in metabolism. Hence, studying individual ROCK isoforms to reveal their specific roles and principal mechanisms in white and beige adipogenesis, insulin sensitivity, energy balancing regulation, and obesity development will facilitate significant breakthroughs for systemic treatment with isoform-selective inhibitors. In this review, we give an overview of ROCK functions in the pathogenesis of obesity and insulin resistance with a particular focus on the current understanding of ROCK isoform signaling in white and beige adipogenesis, obesity and thermogenesis in adipose tissue and other major metabolic organs involved in energy homeostasis regulation.
Collapse
Affiliation(s)
- Lei Wei
- *Correspondence: Lei Wei, ; Jianjian Shi,
| | | |
Collapse
|
10
|
Weber AJ, Adamson AB, Greathouse KM, Andrade JP, Freeman CD, Seo JV, Rae RJ, Walker CK, Herskowitz JH. Conditional deletion of ROCK2 induces anxiety-like behaviors and alters dendritic spine density and morphology on CA1 pyramidal neurons. Mol Brain 2021; 14:169. [PMID: 34794469 PMCID: PMC8600782 DOI: 10.1186/s13041-021-00878-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 11/04/2021] [Indexed: 12/25/2022] Open
Abstract
Rho-associated kinase isoform 2 (ROCK2) is an attractive drug target for several neurologic disorders. A critical barrier to ROCK2-based research and therapeutics is the lack of a mouse model that enables investigation of ROCK2 with spatial and temporal control of gene expression. To overcome this, we generated ROCK2fl/fl mice. Mice expressing Cre recombinase in forebrain excitatory neurons (CaMKII-Cre) were crossed with ROCK2fl/fl mice (Cre/ROCK2fl/fl), and the contribution of ROCK2 in behavior as well as dendritic spine morphology in the hippocampus, medial prefrontal cortex (mPFC), and basolateral amygdala (BLA) was examined. Cre/ROCK2fl/fl mice spent reduced time in the open arms of the elevated plus maze and increased time in the dark of the light-dark box test compared to littermate controls. These results indicated that Cre/ROCK2fl/fl mice exhibited anxiety-like behaviors. To examine dendritic spine morphology, individual pyramidal neurons in CA1 hippocampus, mPFC, and the BLA were targeted for iontophoretic microinjection of fluorescent dye, followed by high-resolution confocal microscopy and neuronal 3D reconstructions for morphometry analysis. In dorsal CA1, Cre/ROCK2fl/fl mice displayed significantly increased thin spine density on basal dendrites and reduced mean spine head volume across all spine types on apical dendrites. In ventral CA1, Cre/ROCK2fl/fl mice exhibited significantly increased spine length on apical dendrites. Spine density and morphology were comparable in the mPFC and BLA between both genotypes. These findings suggest that neuronal ROCK2 mediates spine density and morphology in a compartmentalized manner among CA1 pyramidal cells, and that in the absence of ROCK2 these mechanisms may contribute to anxiety-like behaviors.
Collapse
Affiliation(s)
- Audrey J Weber
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, 1825 University Blvd, Birmingham, AL, 35294, USA
| | - Ashley B Adamson
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, 1825 University Blvd, Birmingham, AL, 35294, USA
| | - Kelsey M Greathouse
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, 1825 University Blvd, Birmingham, AL, 35294, USA
| | - Julia P Andrade
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, 1825 University Blvd, Birmingham, AL, 35294, USA
| | - Cameron D Freeman
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, 1825 University Blvd, Birmingham, AL, 35294, USA
| | - Jung Vin Seo
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, 1825 University Blvd, Birmingham, AL, 35294, USA
| | - Rosaria J Rae
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, 1825 University Blvd, Birmingham, AL, 35294, USA
| | - Courtney K Walker
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, 1825 University Blvd, Birmingham, AL, 35294, USA
| | - Jeremy H Herskowitz
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, 1825 University Blvd, Birmingham, AL, 35294, USA.
| |
Collapse
|
11
|
Singh A, Behl T, Sehgal A, Singh S, Sharma N, Mani V, Alsubayiel AM, Bhatia S, Al-Harrasi A, Bungau S. Exploring the therapeutic promise of targeting Rho kinase in rheumatoid arthritis. Inflammopharmacology 2021; 29:1641-1651. [PMID: 34704172 DOI: 10.1007/s10787-021-00884-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 10/10/2021] [Indexed: 01/28/2023]
Abstract
Rheumatoid arthritis (RA) is a prevalent systemic autoimmune disease caused by dysregulated inflammatory reactions, T lymphocyte invasion into the joints, and articular thickening. Immune cells, primarily tumor necrosis factor-alpha (TNF-α) and chemokines (interleukin or IL-1), which are predominantly generated by activated macrophages cells, have also been involved with the pathogenesis of rheumatoid arthritis. Rho GTPases are integral factors of biochemical cascades utilized by antigens, and also by cellular receptors, cytokines, and chemokines, to modulate inflammatory reactions, according to growing data. The Rho family is a group of G proteins that govern a variety of biological and physiological activities such as mobility, actin stress fiber production, growth, and polarity. Research suggests that the Rho A and Rho-associated coiled-coil kinase (ROCK) regulatory cascade could be essential in several autoimmune conditions, including RA. ROCK is activated in the synovial of rheumatoid arthritis patients, while the blocking of ROCK with fasudil could also decrease IL-6, TNF-α, and IL-1. This review covers current developments in understanding the overactivation of Rho enzyme activity in RA suppressed by ROCK inhibitors which can be utilized for the treatment of autoimmune disease. We offer an outline of the function of ROCK inhibitors in immune cells and discuss findings which emphasize the rising participation of this category of kinases within the pathological process of autoimmune disorders. Assuming the potential ability of ROCK as a therapeutic, we define approaches that might be used to inhibit Rho kinase activity in rheumatoid disorders.
Collapse
Affiliation(s)
- Anuja Singh
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India.
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India
| | - Vasudevan Mani
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah, Saudi Arabia
| | - Amal M Alsubayiel
- Department of Pharmaceutics, College of Pharmacy, Qassim University, Buraydah, Saudi Arabia
| | - Saurabh Bhatia
- Natural and Medical Sciences Research Centre, University of Nizwa, Nizwa, Oman.,School of Health Science, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Centre, University of Nizwa, Nizwa, Oman
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
12
|
Gissler MC, Anto-Michel N, Pennig J, Scherrer P, Li X, Marchini T, Pfeiffer K, Härdtner C, Abogunloko T, Mwinyella T, Sol Mitre L, Spiga L, Koentges C, Smolka C, von Elverfeldt D, Hoppe N, Stachon P, Dufner B, Heidt T, Piepenburg S, Hilgendorf I, Bjune JI, Dankel SN, Mellgren G, Seifert G, Eisenhardt SU, Bugger H, von Zur Muhlen C, Bode C, Zirlik A, Wolf D, Willecke F. Genetic Deficiency of TRAF5 Promotes Adipose Tissue Inflammation and Aggravates Diet-Induced Obesity in Mice. Arterioscler Thromb Vasc Biol 2021; 41:2563-2574. [PMID: 34348490 DOI: 10.1161/atvbaha.121.316677] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective: The accumulation of inflammatory leukocytes is a prerequisite of adipose tissue inflammation during cardiometabolic disease. We previously reported that a genetic deficiency of the intracellular signaling adaptor TRAF5 (TNF [tumor necrosis factor] receptor-associated factor 5) accelerates atherosclerosis in mice by increasing inflammatory cell recruitment. Here, we tested the hypothesis that an impairment of TRAF5 signaling modulates adipose tissue inflammation and its metabolic complications in a model of diet-induced obesity in mice. Approach and Results: To induce diet-induced obesity and adipose tissue inflammation, wild-type or Traf5-/- mice consumed a high-fat diet for 18 weeks. Traf5-/- mice showed an increased weight gain, impaired insulin tolerance, and increased fasting blood glucose. Weight of livers and peripheral fat pads was increased in Traf5-/- mice, whereas lean tissue weight and growth were not affected. Flow cytometry of the stromal vascular fraction of visceral adipose tissue from Traf5-/- mice revealed an increase in cytotoxic T cells, CD11c+ macrophages, and increased gene expression of proinflammatory cytokines and chemokines. At the level of cell types, expression of TNF[alpha], MIP (macrophage inflammatory protein)-1[alpha], MCP (monocyte chemoattractant protein)-1, and RANTES (regulated on activation, normal T-cell expressed and secreted) was significantly upregulated in Traf5-deficient adipocytes but not in Traf5-deficient leukocytes from visceral adipose tissue. Finally, Traf5 expression was lower in adipocytes from obese patients and mice and recovered in adipose tissue of obese patients one year after bariatric surgery. Conclusions: We show that a genetic deficiency of TRAF5 in mice aggravates diet-induced obesity and its metabolic derangements by a proinflammatory response in adipocytes. Our data indicate that TRAF5 may promote anti-inflammatory and obesity-preventing signaling events in adipose tissue.
Collapse
Affiliation(s)
- Mark Colin Gissler
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Nathaly Anto-Michel
- Department of Cardiology, Medical University of Graz, Austria (N.A.M., H.B., A.Z.)
| | - Jan Pennig
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Philipp Scherrer
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Xiaowei Li
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Timoteo Marchini
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Katharina Pfeiffer
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Carmen Härdtner
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Tijani Abogunloko
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Timothy Mwinyella
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Lucia Sol Mitre
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Lisa Spiga
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Christoph Koentges
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
- Institute of Neuropathology (C.K.), Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Christian Smolka
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Dominik von Elverfeldt
- Department of Radiology, Medical Physics (D.v.E.), Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Natalie Hoppe
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Peter Stachon
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Bianca Dufner
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Timo Heidt
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Sven Piepenburg
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Ingo Hilgendorf
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Jan-Inge Bjune
- Center for Diabetes Research (J.-I.B., S.N.D., G.M.), University of Bergen, Norway
- Mohn Nutrition Research Laboratory, Department of Clinical Science (J.-I.B., S.N.D., G.M.), University of Bergen, Norway
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway (J.-I.B., S.N.D., G.M.)
| | - Simon N Dankel
- Center for Diabetes Research (J.-I.B., S.N.D., G.M.), University of Bergen, Norway
- Mohn Nutrition Research Laboratory, Department of Clinical Science (J.-I.B., S.N.D., G.M.), University of Bergen, Norway
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway (J.-I.B., S.N.D., G.M.)
| | - Gunnar Mellgren
- Center for Diabetes Research (J.-I.B., S.N.D., G.M.), University of Bergen, Norway
- Mohn Nutrition Research Laboratory, Department of Clinical Science (J.-I.B., S.N.D., G.M.), University of Bergen, Norway
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway (J.-I.B., S.N.D., G.M.)
| | - Gabriel Seifert
- Department of General and Visceral Surgery (G.S.), Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Steffen U Eisenhardt
- Department of Plastic and Hand Surgery, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Breisgau, Germany (S.U.E.)
| | - Heiko Bugger
- Department of Cardiology, Medical University of Graz, Austria (N.A.M., H.B., A.Z.)
| | - Constantin von Zur Muhlen
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Christoph Bode
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Andreas Zirlik
- Department of Cardiology, Medical University of Graz, Austria (N.A.M., H.B., A.Z.)
| | - Dennis Wolf
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
| | - Florian Willecke
- Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Germany (M.C.G., J.P., P.S., X.L., T. Marchini, K.P., C.H., T.A., T. Mwinyella, L.S.M., L.S., C.K., C.S., N.H., P.S., B.D., T.H., S.P., I.H., C.v.z.M., C.B., D.W., F.W.)
- Clinic for General and Interventional Cardiology/Angiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany (F.W.)
| |
Collapse
|
13
|
Tran NNQ, Chun KH. ROCK2-Specific Inhibitor KD025 Suppresses Adipocyte Differentiation by Inhibiting Casein Kinase 2. Molecules 2021; 26:4747. [PMID: 34443331 PMCID: PMC8401933 DOI: 10.3390/molecules26164747] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/30/2021] [Accepted: 08/02/2021] [Indexed: 11/16/2022] Open
Abstract
KD025, a ROCK2 isoform-specific inhibitor, has an anti-adipogenic activity which is not mediated by ROCK2 inhibition. To identify the target, we searched binding targets of KD025 by using the KINOMEscanTM screening platform, and we identified casein kinase 2 (CK2) as a novel target. KD025 showed comparable binding affinity to CK2α (Kd = 128 nM). By contrast, CK2 inhibitor CX-4945 and ROCK inhibitor fasudil did not show such cross-reactivity. In addition, KD025 effectively inhibited CK2 at a nanomolar concentration (IC50 = 50 nM). We examined if the inhibitory effect of KD025 on adipocyte differentiation is through the inhibition of CK2. Both CX-4945 and KD025 suppressed the generation of lipid droplets and the expression of proadipogenic genes Pparg and Cebpa in 3T3-L1 cells during adipocyte differentiation. Fasudil exerted no significant effect on the quantity of lipid droplets, but another ROCK inhibitor Y-27632 increased the expression of Pparg and Cebpa. Both CX-4945 and KD025 acted specifically in the middle stage (days 1-3) but were ineffective when treated at days 0-1 or the late stages, indicating that CX-4945 and KD025 may regulate the same target, CK2. The mRNA and protein levels of CK2α and CK2β generally decreased in 3T3-L1 cells at day 2 but recovered thereafter. Other well-known CK2 inhibitors DMAT and quinalizarin inhibited effectively the differentiation of 3T3-L1 cells. Taken together, the results of this study confirmed that KD025 inhibits ROCK2 and CK2, and that the inhibitory effect on adipocyte differentiation is through the inhibition of CK2.
Collapse
Affiliation(s)
| | - Kwang-Hoon Chun
- Gachon Institute of Pharmaceutical Sciences, College of Pharmacy, Gachon University, Incheon 21936, Korea;
| |
Collapse
|
14
|
Muñoz VR, Gaspar RC, Severino MB, Macêdo APA, Simabuco FM, Ropelle ER, Cintra DE, da Silva ASR, Kim YB, Pauli JR. Exercise Counterbalances Rho/ROCK2 Signaling Impairment in the Skeletal Muscle and Ameliorates Insulin Sensitivity in Obese Mice. Front Immunol 2021; 12:702025. [PMID: 34234788 PMCID: PMC8256841 DOI: 10.3389/fimmu.2021.702025] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 06/07/2021] [Indexed: 11/29/2022] Open
Abstract
Physical exercise is considered a fundamental strategy in improving insulin sensitivity and glucose uptake in skeletal muscle. However, the molecular mechanisms underlying this regulation, primarily on skeletal muscle glucose uptake, are not fully understood. Recent evidence has shown that Rho-kinase (ROCK) isoforms play a pivotal role in regulating skeletal muscle glucose uptake and systemic glucose homeostasis. The current study evaluated the effect of physical exercise on ROCK2 signaling in skeletal muscle of insulin-resistant obese animals. Physiological (ITT) and molecular analysis (immunoblotting, and RT-qPCR) were performed. The contents of RhoA and ROCK2 protein were decreased in skeletal muscle of obese mice compared to control mice but were restored to normal levels in response to physical exercise. The exercised animals also showed higher phosphorylation of insulin receptor substrate 1 (IRS1 Serine 632/635) and protein kinase B (Akt) in the skeletal muscle. However, phosphatase and tensin homolog (PTEN) and protein-tyrosine phosphatase-1B (PTP-1B), both inhibitory regulators for insulin action, were increased in obesity but decreased after exercise. The impact of ROCK2 action on muscle insulin signaling is further underscored by the fact that impaired IRS1 and Akt phosphorylation caused by palmitate in C2C12 myotubes was entirely restored by ROCK2 overexpression. These results suggest that the exercise-induced upregulation of RhoA-ROCK2 signaling in skeletal muscle is associated with increased systemic insulin sensitivity in obese mice and further implicate that muscle ROCK2 could be a potential target for treating obesity-linked metabolic disorders.
Collapse
Affiliation(s)
- Vitor R Muñoz
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, Brazil
| | - Rafael C Gaspar
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, Brazil
| | - Matheus B Severino
- Multidisciplinary Laboratory of Food and Health, School of Applied Sciences, State University of Campinas, Limeira, Brazil
| | - Ana P A Macêdo
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, Brazil
| | - Fernando M Simabuco
- Multidisciplinary Laboratory of Food and Health, School of Applied Sciences, State University of Campinas, Limeira, Brazil
| | - Eduardo R Ropelle
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, Brazil
| | - Dennys E Cintra
- Laboratory of Nutritional Genomics, University of Campinas (UNICAMP), Limeira, Brazil
| | - Adelino S R da Silva
- Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, Brazil.,School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Young-Bum Kim
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - José Rodrigo Pauli
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, Brazil
| |
Collapse
|
15
|
Wen YT, Huang CW, Liu CP, Chen CH, Tu CM, Hwang CS, Chen YH, Chen WR, Lin KL, Ho YC, Chen TC, Tsai RK. Inhibition of Retinal Ganglion Cell Loss By a Novel ROCK Inhibitor (E212) in Ischemic Optic Nerve Injury Via Antioxidative and Anti-Inflammatory Actions. Invest Ophthalmol Vis Sci 2021; 62:21. [PMID: 34015079 PMCID: PMC8142697 DOI: 10.1167/iovs.62.6.21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 03/24/2021] [Indexed: 11/24/2022] Open
Abstract
Purpose This study investigated the neuroprotective effects of administration of ROCK inhibitor E212 on ischemic optic neuropathy. Methods Rats received an intravitreal injection of either E212 or PBS immediately after optic nerve infarct. The oxidative stress in the retina was detected by performing superoxide dismutase activity and CellROX assays. The integrity of retinal pigment epithelium was determined by staining of zona occludens 1. The visual function, retinal ganglion cell (RGC) density, and RGC apoptosis were determined by using flash visual-evoked potential analysis, retrograde FluoroGold labeling, and TdT-dUTP nick end-labeling assay. Macrophage infiltration was detected by staining for ED1. The protein levels of TNF-α, p-CRMP, p-AKT1, p-STAT3, and CD206 were evaluated using Western blotting. Results Administration of E212 resulted in a 1.23-fold increase in the superoxide dismutase activity of the retina and 2.28-fold decrease in RGC-produced reactive oxygen species as compared to the levels observed upon treatment with PBS (P < 0.05). Moreover, E212 prevented the disruption of the blood-retinal barrier (BRB) in contrast to PBS. The P1-N2 amplitude and RGC density in the E212-treated group were 1.75- and 2.05-fold higher, respectively, than those in the PBS-treated group (P < 0.05). The numbers of apoptotic RGCs and macrophages were reduced by 2.93- and 2.54-fold, respectively, in the E212-treated group compared with those in the PBS-treated group (P < 0.05). The levels of p-AKT1, p-STAT3, and CD206 were increased, whereas those of p-PTEN, p-CRMP2, and TNF-α were decreased after treatment with E212 (P < 0.05). Conclusions Treatment with E212 suppresses oxidative stress, BRB disruption, and neuroinflammation to protect the visual function in ischemic optic neuropathy.
Collapse
Affiliation(s)
- Yao-Tseng Wen
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Ching-Wen Huang
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chih-Peng Liu
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Chih-Hung Chen
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Chia-Mu Tu
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Chrong-Shiong Hwang
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Yi-Hsun Chen
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Wan-Ru Chen
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Keh-Liang Lin
- Department of Medical laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | - Yu-Chieh Ho
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Ta-Ching Chen
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Rong-Kung Tsai
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
- Doctoral Degree Program in Translational Medicine, Tzu Chi University and Academia Sinica, Hualien, Taiwan
| |
Collapse
|
16
|
Machin PA, Tsonou E, Hornigold DC, Welch HCE. Rho Family GTPases and Rho GEFs in Glucose Homeostasis. Cells 2021; 10:cells10040915. [PMID: 33923452 PMCID: PMC8074089 DOI: 10.3390/cells10040915] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/11/2021] [Accepted: 04/13/2021] [Indexed: 12/17/2022] Open
Abstract
Dysregulation of glucose homeostasis leading to metabolic syndrome and type 2 diabetes is the cause of an increasing world health crisis. New intriguing roles have emerged for Rho family GTPases and their Rho guanine nucleotide exchange factor (GEF) activators in the regulation of glucose homeostasis. This review summates the current knowledge, focusing in particular on the roles of Rho GEFs in the processes of glucose-stimulated insulin secretion by pancreatic β cells and insulin-stimulated glucose uptake into skeletal muscle and adipose tissues. We discuss the ten Rho GEFs that are known so far to regulate glucose homeostasis, nine of which are in mammals, and one is in yeast. Among the mammalian Rho GEFs, P-Rex1, Vav2, Vav3, Tiam1, Kalirin and Plekhg4 were shown to mediate the insulin-stimulated translocation of the glucose transporter GLUT4 to the plasma membrane and/or insulin-stimulated glucose uptake in skeletal muscle or adipose tissue. The Rho GEFs P-Rex1, Vav2, Tiam1 and β-PIX were found to control the glucose-stimulated release of insulin by pancreatic β cells. In vivo studies demonstrated the involvement of the Rho GEFs P-Rex2, Vav2, Vav3 and PDZ-RhoGEF in glucose tolerance and/or insulin sensitivity, with deletion of these GEFs either contributing to the development of metabolic syndrome or protecting from it. This research is in its infancy. Considering that over 80 Rho GEFs exist, it is likely that future research will identify more roles for Rho GEFs in glucose homeostasis.
Collapse
Affiliation(s)
- Polly A. Machin
- Signalling Programme, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK; (P.A.M.); (E.T.)
| | - Elpida Tsonou
- Signalling Programme, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK; (P.A.M.); (E.T.)
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Cambridge CB22 3AT, UK;
| | - David C. Hornigold
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Cambridge CB22 3AT, UK;
| | - Heidi C. E. Welch
- Signalling Programme, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK; (P.A.M.); (E.T.)
- Correspondence: ; Tel.: +44-(0)1223-496-596
| |
Collapse
|
17
|
Weber AJ, Herskowitz JH. Perspectives on ROCK2 as a Therapeutic Target for Alzheimer's Disease. Front Cell Neurosci 2021; 15:636017. [PMID: 33790742 PMCID: PMC8005730 DOI: 10.3389/fncel.2021.636017] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/19/2021] [Indexed: 12/12/2022] Open
Abstract
Rho-associated coiled-coil containing kinase isoform 2 (ROCK2) is a member of the AGC family of serine/threonine kinases and an extensively studied regulator of actin-mediated cytoskeleton contractility. Over the past decade, new evidence has emerged that suggests ROCK2 regulates autophagy. Recent studies indicate that dysregulation of autophagy contributes to the development of misfolded tau aggregates among entorhinal cortex (EC) excitatory neurons in early Alzheimer's disease (AD). While the accumulation of tau oligomers and fibrils is toxic to neurons, autophagy facilitates the degradation of these pathologic species and represents a major cellular pathway for tau disposal in neurons. ROCK2 is expressed in excitatory neurons and pharmacologic inhibition of ROCK2 can induce autophagy pathways. In this mini-review, we explore potential mechanisms by which ROCK2 mediates autophagy and actin dynamics and discuss how these pathways represent therapeutic avenues for Alzheimer's disease.
Collapse
Affiliation(s)
| | - Jeremy H. Herskowitz
- Center for Neurodegeneration and Experimental Therapeutics, Departments of Neurology and Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
18
|
Özdemİr A, Ark M. A novel ROCK inhibitor: off-target effects of metformin. ACTA ACUST UNITED AC 2021; 45:35-45. [PMID: 33597820 PMCID: PMC7877715 DOI: 10.3906/biy-2004-12] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 11/10/2020] [Indexed: 12/14/2022]
Abstract
In drug discovery, most small molecules cannot cross many stages, only a few can become drug candidates. Once the drug molecule is approved and marketed, nontarget effects that are not easily distinguishable from the actual target of the drugs might be evaluated. This situation restricts the treatment. Thus, the discovery of new drugs is a very long and expensive process. In recent years, without developing new drugs, the approach of using different and new target molecules in new indications apart from the indications of licensed drug molecules has gained importance.In this study, using the connectivity map program, it was determined that metformin and tolbutamide used in the treatment of type II diabetes had the potential to inhibit Rho kinase. In the experimental results to confirm this data, it has been shown that metformin and tolbutamide decrease the cell area within 24 h and metformin inhibits the activation of Rho kinase in MCF-7 cells.These results indicate that metformin, which is used in the treatment of type II diabetes, acts as a ROCK inhibitor. Metformin has potential in the treatment of various pathological conditions in which Rho kinase has a role.
Collapse
Affiliation(s)
- Aysun Özdemİr
- Department of Pharmacology, Faculty of Pharmacy, Gazi University, Ankara Turkey
| | - Mustafa Ark
- Department of Pharmacology, Faculty of Pharmacy, Gazi University, Ankara Turkey
| |
Collapse
|
19
|
Gupta P, Taiyab A, Hassan MI. Emerging role of protein kinases in diabetes mellitus: From mechanism to therapy. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2020; 124:47-85. [PMID: 33632470 DOI: 10.1016/bs.apcsb.2020.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
Diabetes mellitus has emerged as a severe burden on the medical health system across the globe. Presently, around 422 million people are suffering from diabetes which is speculated to be expanded to about 600 million by 2035. Patients with type 2 diabetes are at increased risk of developing detrimental metabolic and cardiovascular complications. The scientific understanding of this chronic disease and its underlying root cause is not yet fully unraveled. Protein kinases are well known to regulate almost every cellular process through phosphorylation of target protein in diverse signaling pathways. The important role of several protein kinases including AMP-activated protein kinase, IκB kinase and protein kinase C have been well demonstrated in various animal models. They modulate glucose tolerance, inflammation and insulin resistance in the cells via acting on diverse downstream targets and signaling pathways. Thus, modulating the activity of potential human kinases which are significantly involved in diabetes by targeting with small molecule inhibitors could be an attractive therapeutic strategy to tackle diabetes. In this chapter, we have discussed the potential role of protein kinases in glucose metabolism and insulin sensitivity, and in the pathogenesis of diabetes mellitus. Furthermore, the small molecules reported in the literature that can be potentially used for the treatment of diabetes have been discussed in detail.
Collapse
Affiliation(s)
- Preeti Gupta
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Aaliya Taiyab
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India.
| |
Collapse
|
20
|
Vav2 catalysis-dependent pathways contribute to skeletal muscle growth and metabolic homeostasis. Nat Commun 2020; 11:5808. [PMID: 33199701 PMCID: PMC7669868 DOI: 10.1038/s41467-020-19489-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 10/16/2020] [Indexed: 12/17/2022] Open
Abstract
Skeletal muscle promotes metabolic balance by regulating glucose uptake and the stimulation of multiple interorgan crosstalk. We show here that the catalytic activity of Vav2, a Rho GTPase activator, modulates the signaling output of the IGF1- and insulin-stimulated phosphatidylinositol 3-kinase pathway in that tissue. Consistent with this, mice bearing a Vav2 protein with decreased catalytic activity exhibit reduced muscle mass, lack of proper insulin responsiveness and, at much later times, a metabolic syndrome-like condition. Conversely, mice expressing a catalytically hyperactive Vav2 develop muscle hypertrophy and increased insulin responsiveness. Of note, while hypoactive Vav2 predisposes to, hyperactive Vav2 protects against high fat diet-induced metabolic imbalance. These data unveil a regulatory layer affecting the signaling output of insulin family factors in muscle. Skeletal muscle plays a key role in regulating systemic glucose and metabolic homeostasis. Here, the authors show that the catalytic activity of Vav2, an activator of Rho GTPases, modulates those processes by favoring the responsiveness of this tissue to insulin and related factors.
Collapse
|
21
|
Batista TM, Jayavelu AK, Wewer Albrechtsen NJ, Iovino S, Lebastchi J, Pan H, Dreyfuss JM, Krook A, Zierath JR, Mann M, Kahn CR. A Cell-Autonomous Signature of Dysregulated Protein Phosphorylation Underlies Muscle Insulin Resistance in Type 2 Diabetes. Cell Metab 2020; 32:844-859.e5. [PMID: 32888406 PMCID: PMC7875546 DOI: 10.1016/j.cmet.2020.08.007] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/26/2020] [Accepted: 08/11/2020] [Indexed: 02/06/2023]
Abstract
Skeletal muscle insulin resistance is the earliest defect in type 2 diabetes (T2D), preceding and predicting disease development. To what extent this reflects a primary defect or is secondary to tissue cross talk due to changes in hormones or circulating metabolites is unknown. To address this question, we have developed an in vitro disease-in-a-dish model using iPS cells from T2D patients differentiated into myoblasts (iMyos). We find that T2D iMyos in culture exhibit multiple defects mirroring human disease, including an altered insulin signaling, decreased insulin-stimulated glucose uptake, and reduced mitochondrial oxidation. More strikingly, global phosphoproteomic analysis reveals a multidimensional network of signaling defects in T2D iMyos going beyond the canonical insulin-signaling cascade, including proteins involved in regulation of Rho GTPases, mRNA splicing and/or processing, vesicular trafficking, gene transcription, and chromatin remodeling. These cell-autonomous defects and the dysregulated network of protein phosphorylation reveal a new dimension in the cellular mechanisms underlying the fundamental defects in T2D.
Collapse
Affiliation(s)
- Thiago M Batista
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Ashok Kumar Jayavelu
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Nicolai J Wewer Albrechtsen
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany; Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen 2100, Denmark
| | - Salvatore Iovino
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jasmin Lebastchi
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Hui Pan
- Bioinformatics and Biostatistics Core, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jonathan M Dreyfuss
- Bioinformatics and Biostatistics Core, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Anna Krook
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Juleen R Zierath
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Section of Integrative Physiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm 171 76, Sweden
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany; Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - C Ronald Kahn
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
22
|
Abstract
Nonalcoholic hepatitis (NASH) is the progressive inflammatory form of nonalcoholic fatty liver disease. Although the mechanisms of hepatic inflammation in NASH remain incompletely understood, emerging literature implicates the proinflammatory environment created by toxic lipid-induced hepatocyte injury, termed lipotoxicity. Interestingly, numerous NASH-promoting kinases in hepatocytes, immune cells, and adipocytes are activated by the lipotoxic insult associated with obesity. In the current review, we discuss recent advances in NASH-promoting kinases as disease mediators and therapeutic targets. The focus of the review is mainly on the mitogen-activated protein kinases including mixed lineage kinase 3, apoptosis signal-regulating kinase 1, c-Jun N-terminal kinase, and p38 MAPK; the endoplasmic reticulum (ER) stress kinases protein kinase RNA-like ER kinase and inositol-requiring protein-1α; as well as the Rho-associated protein kinase 1. We also discuss various pharmacological agents targeting these stress kinases in NASH that are under different phases of development.
Collapse
Affiliation(s)
- Samar H. Ibrahim
- Division of Gastroenterology & Hepatology in the Department of Pediatrics, Rochester, Minnesota.,Division of Gastroenterology & Hepatology in the Department of Medicine Mayo Clinic, Rochester, Minnesota
| | - Petra Hirsova
- Division of Gastroenterology & Hepatology in the Department of Medicine Mayo Clinic, Rochester, Minnesota
| | - Harmeet Malhi
- Division of Gastroenterology & Hepatology in the Department of Medicine Mayo Clinic, Rochester, Minnesota
| | - Gregory J. Gores
- Division of Gastroenterology & Hepatology in the Department of Medicine Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
23
|
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: 1.8] [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
|
24
|
Guo J, Yang C, Lin Y, Hu G, Wei J, Zhang X, Chen X, Li J. Enhanced peripheral blood miR-324-5p is associated with the risk of metabolic syndrome by suppressing ROCK1. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158727. [PMID: 32353552 DOI: 10.1016/j.bbalip.2020.158727] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 04/17/2020] [Accepted: 04/20/2020] [Indexed: 01/09/2023]
Abstract
The current study aims to evaluate whether peripheral blood miR-324-5p could be used to differentiate patients with metabolic disorders and healthy controls. Our data showed that miR-324-5p levels were elevated in the peripheral blood of patients with hyperglycemia or hyperlipidemia. In addition, the expression of miR-324-5p was enhanced in the peripheral blood and liver of db/db mice. Further study indicated that overexpression of miR-324-5p reduced the activation of the AKT/GSK pathway and increased lipid accumulation, while the inhibition of miR-324-5p activated the AKT/GSK pathway and decreased lipid accumulation. A dual luciferase assay revealed that Rho-associated coiled-coil containing protein kinase 1 (ROCK1) was a target gene of miR-324-5p. In addition, silencing ROCK1 deteriorated lipid and glucose metabolism. More importantly, knockdown of ROCK1 reversed the miR-324-5p inhibitor-induced improvement of glucose and lipid metabolism. In summary, miR-324-5p plays a regulatory role in glucose and lipid metabolism by targeting ROCK1, which is involved in metabolic disorders. The use of miR-324-5p in diagnosing metabolic syndrome is worth investigating and may benefit patients.
Collapse
Affiliation(s)
- Jun Guo
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, PR China
| | - Chunxiao Yang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, PR China; Clinical Laboratory, China Japan Friendship Hospital, PR China
| | - Yajun Lin
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, PR China
| | - Gang Hu
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, PR China
| | - Jie Wei
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, PR China
| | - Xin Zhang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, PR China
| | - Xiehui Chen
- Department of Geriatrics Cardiovascular Medicine, FuWai Hospital, Chinese Academy of Medical Sciences, No. 12, LangShan Road, ShenZhen City, Guangdong Province 518112, PR China.
| | - Jian Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, PR China.
| |
Collapse
|
25
|
Siddiqui MR, Akhtar S, Shahid M, Tauseef M, McDonough K, Shanley TP. miR-144-mediated Inhibition of ROCK1 Protects against LPS-induced Lung Endothelial Hyperpermeability. Am J Respir Cell Mol Biol 2020; 61:257-265. [PMID: 30811958 DOI: 10.1165/rcmb.2018-0235oc] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Dysfunctional endothelial cell (EC) barrier and increased lung vascular permeability is a cardinal feature of acute lung injury and sepsis that may result in a pathophysiological condition characterized by alveolar flooding, pulmonary edema, and subsequent hypoxemia. In lung ECs, activation of Rho-associated kinase-1 (ROCK1) phosphorylates myosin light chain (MLC)-associated phosphatase at its inhibitory site, which favors phosphorylation of MLC, stress fiber formation, and hyperpermeability during acute lung injury. The role of microRNA-144 (miR-144) has been well investigated in many human diseases, including cardiac ischemia/reperfusion-induced injury, lung cancer, and lung viral infection; however, its role in pulmonary EC barrier regulation remains obscure. Here, we investigated the miR-144-mediated mechanism in the protection of endothelial barrier function in an LPS-induced lung injury model. By using transendothelial electrical resistance and transwell permeability assay to examine in vitro permeability and immunofluorescence microscopy to determine barrier integrity, we showed that ectopic expression of miR-144 effectively blocked lung EC barrier disruption and hyperpermeability in response to proinflammatory agents. Furthermore, using a gain-and-loss-of-function strategy, overexpression of miR-144 significantly decreased ROCK1 expression. Concomitantly, miR-144 inhibits ROCK1-mediated phosphorylation of MLC phosphataseThr853 and thus phosphorylation of MLCThr18/Ser19 to counteract stress fiber formation in LPS-activated EC. Finally, in LPS-challenged mice, intranasal delivery of miR-144 mimic via liposomes attenuated endotoxemia-induced increases in lung wet/dry ratio, vascular permeability, and inflammation. In conclusion, these data suggest that miR-144-attenuated activation of inflammatory ROCK1/MLC pathway in vascular ECs is a promising therapeutic strategy to counter inflammatory lung injury.
Collapse
Affiliation(s)
- M Rizwan Siddiqui
- 1Department of Pediatrics, Northwestern University Feinberg School of Medicine and Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,2Stanley Manne Children's Research Institute, Chicago, Illinois; and
| | - Suhail Akhtar
- 1Department of Pediatrics, Northwestern University Feinberg School of Medicine and Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Mohd Shahid
- 3College of Pharmacy, Chicago State University, Chicago, Illinois
| | - Mohammad Tauseef
- 3College of Pharmacy, Chicago State University, Chicago, Illinois
| | - Kelli McDonough
- 1Department of Pediatrics, Northwestern University Feinberg School of Medicine and Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,2Stanley Manne Children's Research Institute, Chicago, Illinois; and
| | - Thomas P Shanley
- 1Department of Pediatrics, Northwestern University Feinberg School of Medicine and Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,2Stanley Manne Children's Research Institute, Chicago, Illinois; and
| |
Collapse
|
26
|
Porazinski S, Parkin A, Pajic M. Rho-ROCK Signaling in Normal Physiology and as a Key Player in Shaping the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1223:99-127. [PMID: 32030687 DOI: 10.1007/978-3-030-35582-1_6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The Rho-ROCK signaling network has a range of specialized functions of key biological importance, including control of essential developmental processes such as morphogenesis and physiological processes including homeostasis, immunity, and wound healing. Deregulation of Rho-ROCK signaling actively contributes to multiple pathological conditions, and plays a major role in cancer development and progression. This dynamic network is critical in modulating the intricate communication between tumor cells, surrounding diverse stromal cells and the matrix, shaping the ever-changing microenvironment of aggressive tumors. In this chapter, we overview the complex regulation of the Rho-ROCK signaling axis, its role in health and disease, and analyze progress made with key approaches targeting the Rho-ROCK pathway for therapeutic benefit. Finally, we conclude by outlining likely future trends and key questions in the field of Rho-ROCK research, in particular surrounding Rho-ROCK signaling within the tumor microenvironment.
Collapse
Affiliation(s)
- Sean Porazinski
- Personalised Cancer Therapeutics Lab, The Kinghorn Cancer Centre, Sydney, NSW, Australia.,Faculty of Medicine, St Vincent's Clinical School, University of NSW, Sydney, NSW, Australia
| | - Ashleigh Parkin
- Personalised Cancer Therapeutics Lab, The Kinghorn Cancer Centre, Sydney, NSW, Australia
| | - Marina Pajic
- Personalised Cancer Therapeutics Lab, The Kinghorn Cancer Centre, Sydney, NSW, Australia. .,Faculty of Medicine, St Vincent's Clinical School, University of NSW, Sydney, NSW, Australia.
| |
Collapse
|
27
|
Muñoz VR, Gaspar RC, Esteca MV, Baptista IL, Vieira RFL, da Silva ASR, de Moura LP, Cintra DE, Ropelle ER, Pauli JR. Physical exercise increases ROCK activity in the skeletal muscle of middle-aged rats. Mech Ageing Dev 2020; 186:111213. [PMID: 32032622 DOI: 10.1016/j.mad.2020.111213] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/17/2020] [Accepted: 01/30/2020] [Indexed: 12/23/2022]
Abstract
The physical exercise is a potential strategy to control age-related metabolic disorders, such as insulin resistance, impaired glucose homeostasis, and type 2 diabetes. Rho-kinase (ROCK) increases skeletal muscle glucose uptake through Insulin Receptor Substrate 1 (IRS1) phosphorylation. Here, we investigated the role of physical exercise in ROCK pathway in the skeletal muscle of Fischer middle-aged rats. Firstly, we observed the ROCK distribution in different skeletal muscle fiber types. ROCK signaling pathway (ROCK1 and ROCK2) and activity (pMYPT1) were higher in the soleus, which was associated with increased insulin signaling pathway (pIR, pIRS1, pPDK, pGSK3β). Middle-aged rats submitted to physical exercise, showed the upregulation of ROCK2 content and normalized RhoA (ROCK activator enzyme) levels in soleus muscle compared with middle-aged sedentary rats. These molecular changes in middle-aged exercised rats were accompanied by higher insulin signaling (pIRS1, pGSK3β, pAS160, GLUT4) in the soleus muscle. Reinforcing these findings, when pharmacological inhibition of ROCK activity was performed (using Y-27632), the insulin signaling pathway and glucose metabolism-related genes (Tpi, Pgk1, Pgam2, Eno3) were decreased in the soleus muscle of exercised rats. In summary, ROCK signaling seems to contribute with whole-body glucose homeostasis (∼50 %) through its higher upregulation in the soleus muscle in middle-aged exercised rats.
Collapse
Affiliation(s)
- Vitor Rosetto Muñoz
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Rafael Calais Gaspar
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Marcos Vinicius Esteca
- Laboratory of Cellular and Tissue Biology, Faculty of Applied Sciences, University of Campinas (UNICAMP), Limeira, Brazil
| | - Igor Luchini Baptista
- Laboratory of Cellular and Tissue Biology, Faculty of Applied Sciences, University of Campinas (UNICAMP), Limeira, Brazil
| | - Renan Fudoli Lins Vieira
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Adelino Sanchez Ramos da Silva
- Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil; School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Leandro Pereira de Moura
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil; OCRC - Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Dennys Esper Cintra
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil; Laboratory of Nutritional Genomics, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Eduardo Rochete Ropelle
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil; OCRC - Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - José Rodrigo Pauli
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil; OCRC - Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.
| |
Collapse
|
28
|
Landry T, Shookster D, Huang H. Tissue-Specific Approaches Reveal Diverse Metabolic Functions of Rho-Kinase 1. Front Endocrinol (Lausanne) 2020; 11:622581. [PMID: 33633690 PMCID: PMC7901932 DOI: 10.3389/fendo.2020.622581] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 12/22/2020] [Indexed: 01/20/2023] Open
Abstract
Rho-kinase 1 (ROCK1) has been implicated in diverse metabolic functions throughout the body, with promising evidence identifying ROCK1 as a therapeutic target in diabetes and obesity. Considering these metabolic roles, several pharmacological inhibitors have been developed to elucidate the mechanisms underlying ROCK1 function. Y27632 and fasudil are two common ROCK1 inhibitors; however, they have varying non-specific selectivity to inhibit other AGC kinase subfamily members and whole-body pharmacological approaches lack tissue-specific insight. As a result, interpretation of studies with these inhibitors is difficult, and alternative approaches are needed to elucidate ROCK1's tissue specific metabolic functions. Fortunately, recent technological advances utilizing molecular carriers or genetic manipulation have facilitated discovery of ROCK1's tissue-specific mechanisms of action. In this article, we review the tissue-specific roles of ROCK1 in the regulation of energy balance and substrate utilization. We highlight prominent metabolic roles in liver, adipose, and skeletal muscle, in which ROCK1 regulates energy expenditure, glucose uptake, and lipid metabolism via inhibition of AMPK2α and paradoxical modulation of insulin signaling. Compared to ROCK1's roles in peripheral tissues, we also describe contradictory functions of ROCK1 in the hypothalamus to increase energy expenditure and decrease food intake via leptin signaling. Furthermore, dysregulated ROCK1 activity in either of these tissues results in metabolic disease phenotypes. Overall, tissue-specific approaches have made great strides in deciphering the many critical metabolic functions of ROCK1 and, ultimately, may facilitate the development of novel treatments for metabolic disorders.
Collapse
Affiliation(s)
- Taylor Landry
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, United States
- Department of Kinesiology, East Carolina University, Greenville, NC, United States
- Human Performance Laboratory, College of Human Performance and Health, East Carolina University, Greenville, NC, United States
| | - Daniel Shookster
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, United States
- Department of Kinesiology, East Carolina University, Greenville, NC, United States
- Human Performance Laboratory, College of Human Performance and Health, East Carolina University, Greenville, NC, United States
| | - Hu Huang
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, United States
- Department of Kinesiology, East Carolina University, Greenville, NC, United States
- Human Performance Laboratory, College of Human Performance and Health, East Carolina University, Greenville, NC, United States
- Department of Physiology, East Carolina University, Greenville, NC, United States
- *Correspondence: Hu Huang,
| |
Collapse
|
29
|
Dankel SN, Røst TH, Kulyté A, Fandalyuk Z, Skurk T, Hauner H, Sagen JV, Rydén M, Arner P, Mellgren G. The Rho GTPase RND3 regulates adipocyte lipolysis. Metabolism 2019; 101:153999. [PMID: 31672447 DOI: 10.1016/j.metabol.2019.153999] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 10/05/2019] [Accepted: 10/23/2019] [Indexed: 01/14/2023]
Abstract
BACKGROUND Adipose tissue plays a crucial role in diet- and obesity-related insulin resistance, with implications for several metabolic diseases. Identification of novel target genes and mechanisms that regulate adipocyte function could lead to improved treatment strategies. RND3 (RhoE/Rho8), a Rho-related GTP-binding protein that inhibits Rho kinase (ROCK) signaling, has been linked to diverse diseases such as apoptotic cardiomyopathy, heart failure, cancer and type 2 diabetes, in part by regulating cytoskeleton dynamics and insulin-mediated glucose uptake. RESULTS We here investigated the expression of RND3 in adipose tissue in human obesity, and discovered a role for RND3 in regulating adipocyte metabolism. In cross-sectional and prospective studies, we observed 5-fold increased adipocyte levels of RND3 mRNA in obesity, reduced levels after surgery-induced weight loss, and positive correlations of RND3 mRNA with adipocyte size and surrogate measures of insulin resistance (HOMA2-IR and circulating triglyceride/high-density lipoprotein cholesterol (TAG/HDL-C) ratio). By screening for RND3-dependent gene expression following siRNA-mediated RND3 knockdown in differentiating human adipocytes, we found downregulation of inflammatory genes and upregulation of genes related to adipocyte ipolysis and insulin signaling. Treatment of adipocytes with tumor necrosis factor alpha (TNFα), lipopolysaccharide (LPS), hypoxia or cAMP analogs increased RND3 mRNA levels 1.5-2-fold. Functional assays in primary human adipocytes confirmed that RND3 knockdown reduces cAMP- and isoproterenol-induced lipolysis, which were mimicked by treating cells with ROCK inhibitor. This effect could partly be explained by reduced protein expression of adipose triglyceride lipase (ATGL) and phosphorylated hormone-sensitive lipase (HSL). CONCLUSION We here uncovered a novel differential expression of adipose RND3 in obesity and insulin resistance, which may at least partly depend on a causal effect of RND3 on adipocyte lipolysis.
Collapse
Affiliation(s)
- Simon N Dankel
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway; Hormone Laboratory, Haukeland University Hospital, N-5021 Bergen, Norway.
| | - Therese H Røst
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway; Hormone Laboratory, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Agné Kulyté
- Department of Medicine (H7), Karolinska Institutet, C2-94 Karolinska University Hospital, Huddinge, 141 86 Stockholm, Sweden
| | - Zina Fandalyuk
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway
| | - Thomas Skurk
- ZIEL Institute for Food and Health, Technical University of Munich, 85354 Freising, Germany; Else Kroener-Fresenius Centre for Nutritional Medicine, School of Medicine, Technical University of Munich, 80992 Munich, Germany
| | - Hans Hauner
- Else Kroener-Fresenius Centre for Nutritional Medicine, School of Medicine, Technical University of Munich, 80992 Munich, Germany; German Center of Diabetes Research, Helmholtz Center, Munich, Germany
| | - Jørn V Sagen
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway; Hormone Laboratory, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Mikael Rydén
- Department of Medicine (H7), Karolinska Institutet, C2-94 Karolinska University Hospital, Huddinge, 141 86 Stockholm, Sweden
| | - Peter Arner
- Department of Medicine (H7), Karolinska Institutet, C2-94 Karolinska University Hospital, Huddinge, 141 86 Stockholm, Sweden
| | - Gunnar Mellgren
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway; Hormone Laboratory, Haukeland University Hospital, N-5021 Bergen, Norway.
| |
Collapse
|
30
|
Muñoz VR, Gaspar RC, Minuzzi LG, dos Santos Canciglieri R, da Silva ASR, de Moura LP, Cintra DE, Ropelle ER, Pauli JR. Rho-kinase activity is upregulated in the skeletal muscle of aged exercised rats. Exp Gerontol 2019; 128:110746. [DOI: 10.1016/j.exger.2019.110746] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 09/25/2019] [Accepted: 09/30/2019] [Indexed: 01/30/2023]
|
31
|
Wei L, Surma M, Yang Y, Tersey S, Shi J. ROCK2 inhibition enhances the thermogenic program in white and brown fat tissue in mice. FASEB J 2019; 34:474-493. [PMID: 31914704 DOI: 10.1096/fj.201901174rr] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 10/09/2019] [Accepted: 10/22/2019] [Indexed: 12/20/2022]
Abstract
The RhoA/ROCK-mediated actin cytoskeleton dynamics have been implicated in adipogenesis. The two ROCK isoforms, ROCK1 and ROCK2, are highly homologous. The contribution of ROCK2 to adipogenesis in vivo has not been elucidated. The present study aimed at the in vivo and in vitro roles of ROCK2 in the regulation of adipogenesis and the development of obesity. We performed molecular, histological, and metabolic analyses in ROCK2+/- and ROCK2+/KD mouse models, the latter harboring an allele with a kinase-dead (KD) mutation. Both ROCK2+/- and ROCK2+/KD mouse models showed a lean body mass phenotype during aging, associated with increased amounts of beige cells in subcutaneous white adipose tissue (sWAT) and increased thermogenic gene expression in all fat depots. ROCK2+/- mice on a high-fat diet showed increased energy expenditure accompanying by reduced obesity, and improved insulin sensitivity. In vitro differentiated ROCK2+/- stromal-vascular (SV) cells revealed increased beige adipogenesis associated with increased thermogenic gene expressions. Treatment with a selective ROCK2 inhibitor, KD025, to inhibit ROCK2 activity in differentiated SV cells reproduced the pro-beige phenotype of ROCK2+/- SV cells. In conclusion, ROCK2 activity-mediated actin cytoskeleton dynamics contribute to the inhibition of beige adipogenesis in WAT, and also promotes age-related and diet-induced fat mass gain and insulin resistance.
Collapse
Affiliation(s)
- Lei Wei
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA.,Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Michelle Surma
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yang Yang
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sarah Tersey
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA.,Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jianjian Shi
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
32
|
Greathouse KM, Henderson BW, Gentry EG, Herskowitz JH. Fasudil or genetic depletion of ROCK1 or ROCK2 induces anxiety-like behaviors. Behav Brain Res 2019; 373:112083. [PMID: 31302146 PMCID: PMC6693674 DOI: 10.1016/j.bbr.2019.112083] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 07/10/2019] [Accepted: 07/10/2019] [Indexed: 12/13/2022]
Abstract
Twenty-nine protein kinase inhibitors have been used to treat human diseases. Out of these, two are Rho-associated protein kinase (ROCK) 1 and 2 inhibitors. The ROCKs heavily influence neuronal architecture and structural plasticity, and ROCKs are putative drug targets for various brain disorders. While the pan-ROCK inhibitor Fasudil has been clinically approved to treat hypertension, heart failure, glaucoma, spinal cord injury, and stroke, a barrier to progress on this therapeutic avenue is the lack of experimental comparisons between pharmacologic and genetic manipulation of ROCKs. Our study begins to address this question using parallel approaches to study behavior in mice that were treated with Fasudil or were heterozygous for ROCK1 or ROCK2. Adult mice treated with Fasudil for thirty days displayed reduced time spent in the open arms of the elevated plus maze, whereas activity in the open field was more analogous to mock-treated animals. Both male and female adult ROCK1+/- and ROCK2+/- mice exhibited reduced time spent in open arms of the elevated plus maze compared to littermate controls. However, ROCK1 or ROCK2 heterozygosity did not alter performance in the open field or Y-maze. These results indicate that chronic treatment with Fasudil induces anxiety-like behaviors that are likely the consequence of ROCK1 and/or ROCK2 inhibition. Our findings may have implications for several ongoing clinical trials using Fasudil or other ROCK-based therapeutics.
Collapse
Affiliation(s)
- Kelsey M Greathouse
- Center for Neurodegeneration and Experimental Therapeutics and Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| | - Benjamin W Henderson
- Center for Neurodegeneration and Experimental Therapeutics and Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| | - Erik G Gentry
- Center for Neurodegeneration and Experimental Therapeutics and Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| | - Jeremy H Herskowitz
- Center for Neurodegeneration and Experimental Therapeutics and Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama 35294, United States.
| |
Collapse
|
33
|
Møller LLV, Klip A, Sylow L. Rho GTPases-Emerging Regulators of Glucose Homeostasis and Metabolic Health. Cells 2019; 8:E434. [PMID: 31075957 PMCID: PMC6562660 DOI: 10.3390/cells8050434] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/02/2019] [Accepted: 05/06/2019] [Indexed: 12/11/2022] Open
Abstract
Rho guanosine triphosphatases (GTPases) are key regulators in a number of cellular functions, including actin cytoskeleton remodeling and vesicle traffic. Traditionally, Rho GTPases are studied because of their function in cell migration and cancer, while their roles in metabolism are less documented. However, emerging evidence implicates Rho GTPases as regulators of processes of crucial importance for maintaining metabolic homeostasis. Thus, the time is now ripe for reviewing Rho GTPases in the context of metabolic health. Rho GTPase-mediated key processes include the release of insulin from pancreatic β cells, glucose uptake into skeletal muscle and adipose tissue, and muscle mass regulation. Through the current review, we cast light on the important roles of Rho GTPases in skeletal muscle, adipose tissue, and the pancreas and discuss the proposed mechanisms by which Rho GTPases act to regulate glucose metabolism in health and disease. We also describe challenges and goals for future research.
Collapse
Affiliation(s)
- Lisbeth Liliendal Valbjørn Møller
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, 2100 Copenhagen Oe, Denmark.
| | - Amira Klip
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada.
| | - Lykke Sylow
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, 2100 Copenhagen Oe, Denmark.
| |
Collapse
|
34
|
Cordeiro AV, Silva VRR, Pauli JR, da Silva ASR, Cintra DE, Moura LP, Ropelle ER. The role of sphingosine-1-phosphate in skeletal muscle: Physiology, mechanisms, and clinical perspectives. J Cell Physiol 2018; 234:10047-10059. [PMID: 30523638 DOI: 10.1002/jcp.27870] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 11/15/2018] [Indexed: 12/21/2022]
Abstract
Sphingolipids were discovered more than a century ago and were simply considered as a class of cell membrane lipids for a long time. However, after the discovery of several intracellular functions and their role in the control of many physiological and pathophysiological conditions, these molecules have gained much attention. For instance, the sphingosine-1-phosphate (S1P) is a circulating bioactive sphingolipid capable of triggering strong intracellular reactions through the family of S1P receptors (S1PRs) spread in several cell types and tissues. Recently, the role of S1P in the control of skeletal muscle metabolism, atrophy, regeneration, and metabolic disorders has been widely investigated. In this review, we summarized the knowledge of S1P and its effects in skeletal muscle metabolism, highlighting the role of S1P/S1PRs axis in skeletal muscle regeneration, fatigue, ceramide accumulation, and insulin resistance. Finally, we discussed the physical exercise role in S1P/S1PRs signaling in skeletal muscle cells, and how this nonpharmacological strategy may be prospective for future investigations due to its ability to increase S1P levels.
Collapse
Affiliation(s)
- André V Cordeiro
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Vagner R R Silva
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - José R Pauli
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil.,School of Applied Sciences, Center of Research in Sport Sciences (CEPECE), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Adelino S R da Silva
- Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, USP, Ribeirão Preto, São Paulo, Brazil.,School of Physical Education and Sport of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Dennys E Cintra
- Laboratory of Nutritional Genomics (LabGeN), School of Applied Sciences, University of Campinas, Limeira, São Paulo, Brazil
| | - Leandro P Moura
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil.,School of Applied Sciences, Center of Research in Sport Sciences (CEPECE), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Eduardo R Ropelle
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil.,School of Applied Sciences, Center of Research in Sport Sciences (CEPECE), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil.,Department of Internal Medicine, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| |
Collapse
|
35
|
Greathouse KM, Boros BD, Deslauriers JF, Henderson BW, Curtis KA, Gentry EG, Herskowitz JH. Distinct and complementary functions of rho kinase isoforms ROCK1 and ROCK2 in prefrontal cortex structural plasticity. Brain Struct Funct 2018; 223:4227-4241. [PMID: 30196430 PMCID: PMC6252131 DOI: 10.1007/s00429-018-1748-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 09/04/2018] [Indexed: 12/13/2022]
Abstract
Rho-associated protein kinases (ROCK) 1 and 2 are attractive drug targets for a range of neurologic disorders; however, a critical barrier to ROCK-based therapeutics is ambiguity over whether there are isoform-specific roles for ROCKs in neuronal structural plasticity. Here, we used a genetics approach to address this long-standing question by analyzing both male and female adult ROCK1+/- and ROCK2+/- mice compared to littermate controls. Individual pyramidal neurons in the medial prefrontal cortex (mPFC) were targeted for iontophoretic microinjection of fluorescent dye, followed by high-resolution confocal microscopy and neuronal 3D reconstructions for morphometry analysis. Increased apical and basal dendritic length and intersections were observed in ROCK1+/- but not ROCK2+/- mice. Although dendritic spine densities were comparable among genotypes, apical spine length was decreased in ROCK1+/- but increased in ROCK2+/- mice. Spine head and neck diameter were reduced similarly in ROCK1+/- and ROCK2+/- mice; however, certain spine morphologic subclasses were more affected than others in a genotype-dependent manner. Biochemical analyses of ROCK substrates in synaptic fractions revealed that phosphorylation of LIM kinase and cofilin were reduced in ROCK1+/- and ROCK2+/- mice, while phosphorylation of myosin light chain was decreased exclusively in ROCK1+/- mice. Collectively, these observations implicate ROCK1 as a novel regulatory factor of neuronal dendritic structure and detail distinct and complementary roles of ROCKs in mPFC dendritic spine structure.
Collapse
Affiliation(s)
- Kelsey M Greathouse
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL, 35294, USA
| | - Benjamin D Boros
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL, 35294, USA
| | - Josue F Deslauriers
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL, 35294, USA
| | - Benjamin W Henderson
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL, 35294, USA
| | - Kendall A Curtis
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL, 35294, USA
| | - Erik G Gentry
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL, 35294, USA
| | - Jeremy H Herskowitz
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL, 35294, USA.
| |
Collapse
|
36
|
Huang H, Lee SH, Sousa-Lima I, Kim SS, Hwang WM, Dagon Y, Yang WM, Cho S, Kang MC, Seo JA, Shibata M, Cho H, Belew GD, Bhin J, Desai BN, Ryu MJ, Shong M, Li P, Meng H, Chung BH, Hwang D, Kim MS, Park KS, Macedo MP, White M, Jones J, Kim YB. Rho-kinase/AMPK axis regulates hepatic lipogenesis during overnutrition. J Clin Invest 2018; 128:5335-5350. [PMID: 30226474 DOI: 10.1172/jci63562] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 09/11/2018] [Indexed: 12/24/2022] Open
Abstract
Obesity is a major risk factor for developing nonalcoholic fatty liver disease (NAFLD). NAFLD is the most common form of chronic liver disease and is closely associated with insulin resistance, ultimately leading to cirrhosis and hepatocellular carcinoma. However, knowledge of the intracellular regulators of obesity-linked fatty liver disease remains incomplete. Here we showed that hepatic Rho-kinase 1 (ROCK1) drives obesity-induced steatosis in mice through stimulation of de novo lipogenesis. Mice lacking ROCK1 in the liver were resistant to diet-induced obesity owing to increased energy expenditure and thermogenic gene expression. Constitutive expression of hepatic ROCK1 was sufficient to promote adiposity, insulin resistance, and hepatic lipid accumulation in mice fed a high-fat diet. Correspondingly, liver-specific ROCK1 deletion prevented the development of severe hepatic steatosis and reduced hyperglycemia in obese diabetic (ob/ob) mice. Of pathophysiological significance, hepatic ROCK1 was markedly upregulated in humans with fatty liver disease and correlated with risk factors clustering around NAFLD and insulin resistance. Mechanistically, we found that hepatic ROCK1 suppresses AMPK activity and a ROCK1/AMPK pathway is necessary to mediate cannabinoid-induced lipogenesis in the liver. Furthermore, treatment with metformin, the most widely used antidiabetes drug, reduced hepatic lipid accumulation by inactivating ROCK1, resulting in activation of AMPK downstream signaling. Taken together, our findings establish a ROCK1/AMPK signaling axis that regulates de novo lipogenesis, providing a unique target for treating obesity-related metabolic disorders such as NAFLD.
Collapse
Affiliation(s)
- Hu Huang
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA.,Department of Kinesiology and Physiology, East Carolina University, East Carolina Diabetes and Obesity Institute, Greenville, North Carolina, USA
| | - Seung-Hwan Lee
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA.,Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Inês Sousa-Lima
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA.,Centro de Estudos de Doenҫas Crónicas (CEDOC), Chronic Disease Research Center, NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Sang Soo Kim
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Won Min Hwang
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Yossi Dagon
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Won-Mo Yang
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Sungman Cho
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Min-Cheol Kang
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Ji A Seo
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA.,Division of Endocrinology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Munehiko Shibata
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Hyunsoo Cho
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Getachew Debas Belew
- Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, Coimbra, Portugal
| | - Jinhyuk Bhin
- Center for Plant Aging Research and Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea
| | - Bhavna N Desai
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Min Jeong Ryu
- Department of Endocrinology and Metabolism, Chungnam National University School of Medicine, Daejeon, Korea
| | - Minho Shong
- Department of Endocrinology and Metabolism, Chungnam National University School of Medicine, Daejeon, Korea
| | - Peixin Li
- Department of Kinesiology and Physiology, East Carolina University, East Carolina Diabetes and Obesity Institute, Greenville, North Carolina, USA.,Department of Comprehensive Surgery Medical and Health Center Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Hua Meng
- Department of Comprehensive Surgery Medical and Health Center Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Byung-Hong Chung
- Department of Nutrition Science, Diabetes Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Daehee Hwang
- Center for Plant Aging Research and Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea
| | - Min Seon Kim
- Department of Internal Medicine, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea
| | - Kyong Soo Park
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| | - Maria Paula Macedo
- Centro de Estudos de Doenҫas Crónicas (CEDOC), Chronic Disease Research Center, NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Morris White
- Department of Endocrinology, Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - John Jones
- Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, Coimbra, Portugal
| | - Young-Bum Kim
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| |
Collapse
|
37
|
Sánchez-Aguilera P, Diaz-Vegas A, Campos C, Quinteros-Waltemath O, Cerda-Kohler H, Barrientos G, Contreras-Ferrat A, Llanos P. Role of ABCA1 on membrane cholesterol content, insulin-dependent Akt phosphorylation and glucose uptake in adult skeletal muscle fibers from mice. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1863:1469-1477. [PMID: 30254016 DOI: 10.1016/j.bbalip.2018.09.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 08/23/2018] [Accepted: 09/17/2018] [Indexed: 02/07/2023]
Abstract
The ATP-binding cassette transporter A1 (ABCA1) promotes cellular cholesterol efflux, leading to cholesterol binding to the extracellular lipid-free apolipoprotein A-I. ABCA1 regulates lipid content, glucose tolerance and insulin sensitivity in adipose tissue. In skeletal muscle, most GLUT4-mediated glucose transport occurs in the transverse tubule, a system composed by specialized cholesterol-enriched invaginations of the plasma membrane. We have reported that insulin resistant mice have higher cholesterol levels in transverse tubule from adult skeletal muscle. These high levels correlate with decreased GLUT4 trafficking and glucose uptake; however, the role of ABCA1 on skeletal muscle insulin-dependent glucose metabolism remains largely unexplored. Here, we evaluated the functional role of the ABCA1 on insulin-dependent signaling pathways, glucose uptake and cellular cholesterol content in adult skeletal muscle. Male mice were fed for 8 weeks with normal chow diet (NCD) or high fat diet (HFD). Compared to NCD-fed mice, ABCA1 mRNA levels and protein content were lower in muscle homogenates from HFD-fed mice. In Flexor digitorum brevis muscle from NCD-fed mice, shABCA1-RFP in vivo electroporation resulted in 65% reduction of ABCA1 protein content, 1.6-fold increased fiber cholesterol levels, 74% reduction in insulin-dependent Akt (Ser473) phosphorylation, total suppression of insulin-dependent GLUT4 translocation and decreased 2-NBDG uptake compared to fibers electroporated with the scrambled plasmid. Pre-incubation with methyl-β cyclodextrin reestablished both GLUT4 translocation and 2-NBDG transport. Based on the present results, we suggest that decreased ABCA1 contributes to the anomalous cholesterol accumulation and decreased glucose transport displayed by skeletal muscle membranes in the insulin resistant condition.
Collapse
Affiliation(s)
- Pablo Sánchez-Aguilera
- Institute for Research in Dental Sciences, Facultad de Odontología, Universidad de Chile, Chile
| | - Alexis Diaz-Vegas
- Departamento Ciencias Biológicas, Facultad Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | | | | | - Hugo Cerda-Kohler
- Institute for Research in Dental Sciences, Facultad de Odontología, Universidad de Chile, Chile
| | | | - Ariel Contreras-Ferrat
- ACCDiS, Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Chile
| | - Paola Llanos
- Institute for Research in Dental Sciences, Facultad de Odontología, Universidad de Chile, Chile; CEMC, Facultad de Medicina, Universidad de Chile, Chile.
| |
Collapse
|
38
|
Lipina C, Walsh SK, Mitchell SE, Speakman JR, Wainwright CL, Hundal HS. GPR55 deficiency is associated with increased adiposity and impaired insulin signaling in peripheral metabolic tissues. FASEB J 2018; 33:1299-1312. [PMID: 30148676 PMCID: PMC6355063 DOI: 10.1096/fj.201800171r] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Emerging evidence indicates that G-protein coupled receptor 55 (GPR55), a nonclassic receptor of the endocannabinoid system that is activated by L-α-lysophosphatidylinositol and various cannabinoid ligands, may regulate endocrine function and energy metabolism. We examined how GPR55 deficiency and modulation affects insulin signaling in skeletal muscle, adipose tissue, and liver alongside expression analysis of proteins implicated in insulin action and energy metabolism. We show that GPR55-null mice display decreased insulin sensitivity in these tissues, as evidenced by reduced phosphorylation of PKB/Akt and its downstream targets, concomitant with increased adiposity and reduced physical activity relative to wild-type counterparts. Impaired tissue insulin sensitivity coincided with reduced insulin receptor substrate-1 abundance in skeletal muscle, whereas in liver and epididymal fat it was associated with increased expression of the 3-phosphoinoistide lipid phosphatase, phosphatase and tensin homolog. In contrast, GPR55 activation enhanced insulin signaling in cultured skeletal muscle cells, adipocytes, and hepatocytes; this response was negated by receptor antagonists and GPR55 gene silencing in L6 myotubes. Sustained GPR55 antagonism in 3T3-L1 adipocytes enhanced expression of proteins implicated in lipogenesis and promoted triglyceride accumulation. Our findings identify GPR55 as a positive regulator of insulin action and adipogenesis and as a potential therapeutic target for countering obesity-induced metabolic dysfunction and insulin resistance.-Lipina, C., Walsh, S. K., Mitchell, S. E., Speakman, J. R., Wainwright, C. L., Hundal, H. S. GPR55 deficiency is associated with increased adiposity and impaired insulin signaling in peripheral metabolic tissues.
Collapse
Affiliation(s)
- Christopher Lipina
- Division of Cell Signalling and Immunology, Sir James Black Centre, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Sarah K Walsh
- Centre for Cardiometabolic Health Research, Robert Gordon University, Aberdeen, United Kingdom
| | - Sharon E Mitchell
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - John R Speakman
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, United Kingdom.,Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Cherry L Wainwright
- Centre for Cardiometabolic Health Research, Robert Gordon University, Aberdeen, United Kingdom
| | - Harinder S Hundal
- Division of Cell Signalling and Immunology, Sir James Black Centre, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
39
|
Muñoz VR, Gaspar RC, Kuga GK, da Rocha AL, Crisol BM, Botezelli JD, Baptista IL, Mekary RA, da Silva ASR, Cintra DE, de Moura LP, Ropelle ER, Pauli JR. Exercise increases Rho-kinase activity and insulin signaling in skeletal muscle. J Cell Physiol 2018; 233:4791-4800. [DOI: 10.1002/jcp.26278] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 11/14/2017] [Indexed: 12/16/2022]
Affiliation(s)
- Vitor R. Muñoz
- Laboratory of Molecular Biology of Exercise; University of Campinas (UNICAMP); Limeira São Paulo Brazil
| | - Rafael C. Gaspar
- Laboratory of Molecular Biology of Exercise; University of Campinas (UNICAMP); Limeira São Paulo Brazil
| | - Gabriel K. Kuga
- Laboratory of Molecular Biology of Exercise; University of Campinas (UNICAMP); Limeira São Paulo Brazil
| | - Alisson L. da Rocha
- Postgraduate Program in Rehabilitation and Functional Performance,; Ribeirão Preto Medical School, USP; Ribeirão Preto São Paulo Brazil
- School of Physical Education and Sport of Ribeirão Preto; University of São Paulo (USP); Ribeirão Preto São Paulo Brazil
| | - Barbara M. Crisol
- Laboratory of Molecular Biology of Exercise; University of Campinas (UNICAMP); Limeira São Paulo Brazil
| | - José D. Botezelli
- Laboratory of Molecular Biology of Exercise; University of Campinas (UNICAMP); Limeira São Paulo Brazil
| | - Igor L. Baptista
- Laboratory of Cell and Tissue Biology; University of Campinas (UNICAMP); Limeira São Paulo Brazil
| | - Rania A. Mekary
- Department of Nutrition; Harvard T. Chan School of Public Health; Boston Massachusetts
- Department of Pharmaceutical Business and Administrative Sciences; MCPHS University; Boston Massachusetts
| | - Adelino S. R. da Silva
- Laboratory of Cell and Tissue Biology; University of Campinas (UNICAMP); Limeira São Paulo Brazil
| | - Dennys E. Cintra
- Laboratory of Nutritional Genomics; University of Campinas (UNICAMP); Limeira São Paulo Brazil
- OCRC - Obesity and Comorbidities Research Center; University of Campinas (UNICAMP); Campinas São Paulo Brazil
| | - Leandro P. de Moura
- Laboratory of Molecular Biology of Exercise; University of Campinas (UNICAMP); Limeira São Paulo Brazil
- OCRC - Obesity and Comorbidities Research Center; University of Campinas (UNICAMP); Campinas São Paulo Brazil
- CEPECE - Center of Research in Sport Sciences. School of Applied Sciences; University of Campinas (UNICAMP); Limeira São Paulo Brazil
| | - Eduardo R. Ropelle
- Laboratory of Molecular Biology of Exercise; University of Campinas (UNICAMP); Limeira São Paulo Brazil
- OCRC - Obesity and Comorbidities Research Center; University of Campinas (UNICAMP); Campinas São Paulo Brazil
- CEPECE - Center of Research in Sport Sciences. School of Applied Sciences; University of Campinas (UNICAMP); Limeira São Paulo Brazil
| | - José R. Pauli
- Laboratory of Molecular Biology of Exercise; University of Campinas (UNICAMP); Limeira São Paulo Brazil
- OCRC - Obesity and Comorbidities Research Center; University of Campinas (UNICAMP); Campinas São Paulo Brazil
- CEPECE - Center of Research in Sport Sciences. School of Applied Sciences; University of Campinas (UNICAMP); Limeira São Paulo Brazil
| |
Collapse
|
40
|
Seo JA, Kang MC, Ciaraldi TP, Kim SS, Park KS, Choe C, Hwang WM, Lim DM, Farr O, Mantzoros C, Henry RR, Kim YB. Circulating ApoJ is closely associated with insulin resistance in human subjects. Metabolism 2018; 78:155-166. [PMID: 28986164 PMCID: PMC5765540 DOI: 10.1016/j.metabol.2017.09.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 09/19/2017] [Accepted: 09/20/2017] [Indexed: 11/20/2022]
Abstract
OBJECTIVE Insulin resistance is a major risk factor for type 2 diabetes. ApolipoproteinJ (ApoJ) has been implicated in altered pathophysiologic states including cardiovascular and Alzheimer's disease. However, the function of ApoJ in regulation of glucose homeostasis remains unclear. This study sought to determine whether serum ApoJ levels are associated with insulin resistance in human subjects and if they change after interventions that improve insulin sensitivity. METHODS Serum ApoJ levels and insulin resistance status were assessed in nondiabetic (ND) and type 2 diabetic (T2D) subjects. The impacts of rosiglitazone or metformin therapy on serum ApoJ levels and glucose disposal rate (GDR) during a hyperinsulinemic/euglycemic clamp were evaluated in a separate cohort of T2D subjects. Total ApoJ protein or that associated with the HDL and LDL fractions was measured by immunoblotting or ELISA. RESULTS Fasting serum ApoJ levels were greatly elevated in T2D subjects (ND vs T2D; 100±8.3 vs. 150.6±8.5AU, P<0.0001). Circulating ApoJ levels strongly correlated with fasting glucose, fasting insulin, HOMA-IR, and BMI. ApoJ levels were significantly and independently associated with HOMA-IR, even after adjustment for age, sex, and BMI. Rosiglitazone treatment in T2D subjects resulted in a reduction in serum ApoJ levels (before vs. after treatment; 100±13.9 vs. 77±15.2AU, P=0.015), whereas metformin had no effect on ApoJ levels. The change in ApoJ levels during treatment was inversely associated with the change in GDR. Interestingly, ApoJ content in the LDL fraction was inversely associated with HOMA-IR. CONCLUSION Serum ApoJ levels are closely correlated with the magnitude of insulin resistance regardless of obesity, and decrease along with improvement of insulin resistance in response only to rosiglitazone in type 2 diabetes.
Collapse
Affiliation(s)
- Ji A Seo
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States; Division of Endocrinology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Min-Cheol Kang
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States
| | - Theodore P Ciaraldi
- Veterans Affairs San Diego Healthcare System (9111G), San Diego, CA 92161, United States; Department of Medicine, University of California, San Diego, La Jolla, CA 92093, United States
| | - Sang Soo Kim
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States
| | - Kyong Soo Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea; Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| | - Charles Choe
- Veterans Affairs San Diego Healthcare System (9111G), San Diego, CA 92161, United States; Department of Medicine, University of California, San Diego, La Jolla, CA 92093, United States
| | - Won Min Hwang
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States; Division of Nephrology, Department of Internal Medicine, College of Medicine, Konyang University, Daejeon, Korea
| | - Dong Mee Lim
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States; Division of Nephrology, Department of Internal Medicine, College of Medicine, Konyang University, Daejeon, Korea
| | - Olivia Farr
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States
| | - Christos Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States
| | - Robert R Henry
- Veterans Affairs San Diego Healthcare System (9111G), San Diego, CA 92161, United States; Department of Medicine, University of California, San Diego, La Jolla, CA 92093, United States
| | - Young-Bum Kim
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States; Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea.
| |
Collapse
|
41
|
Zheng G, Mo F, Ling C, Peng H, Gu W, Li M, Chen Z. Portulaca oleracea L. alleviates liver injury in streptozotocin-induced diabetic mice. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 12:47-55. [PMID: 29343942 PMCID: PMC5749558 DOI: 10.2147/dddt.s121084] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Purslane is a widespread succulent herb that exhibits various pharmacological effects. The purpose of this study was to evaluate the protective effect of Portulaca oleracea L. (purslane) on streptozotocin-induced diabetes in mice. Oral glucose-tolerance tests were carried out to assess blood glucose levels and body weight and food intake were recorded. The biochemical parameters anti-aspartate aminotransferase, alanine aminotransferase, insulin, triglycerides, total cholesterol, IL-6, IL-1β, and TNFα were also measured. The pathological condition of liver tissues were examined by hematoxylin–eosin staining. Rho, ROCK1, ROCK2, NFκBp65, p-NFκBp65, IκBα, and p-IκBα expression in liver tissue were analyzed by Western blot. Purslane increased body weight and decreased food intake. Purslane also significantly reduced concentrations of glucose, anti-aspartate aminotransferase, alanine aminotransferase, triglycerides, total cholesterol, IL-6, IL-1β, and TNFα in serum. Serum insulin was elevated with purslane treatment. In addition, pathologic liver changes in diabetic mice were also alleviated by purslane. Obtained data revealed that purslane restored the levels of Rho–NFκB signaling-related proteins in comparison with those of diabetic mice. Above all, it can be assumed that purslane might play a positive role in regulating streptozotocin-induced liver injury through suppressing the Rho–NFκB pathway.
Collapse
Affiliation(s)
- Guoyin Zheng
- Department of Traditional Chinese Medicine, Changhai Hospital
| | - Fengfeng Mo
- Department of Military Hygiene, Second Military Medical University
| | - Chen Ling
- Department of Biology, School of Life Science, Fudan University, Shanghai, People's Republic of China
| | - Hao Peng
- Department of Traditional Chinese Medicine, Changhai Hospital
| | - Wei Gu
- Department of Traditional Chinese Medicine, Changhai Hospital
| | - Min Li
- Department of Military Hygiene, Second Military Medical University
| | - Zhe Chen
- Department of Traditional Chinese Medicine, Changhai Hospital
| |
Collapse
|
42
|
Gastebois C, Chanon S, Rome S, Durand C, Pelascini E, Jalabert A, Euthine V, Pialoux V, Blanc S, Simon C, Lefai E. Transition from physical activity to inactivity increases skeletal muscle miR-148b content and triggers insulin resistance. Physiol Rep 2017; 4:4/17/e12902. [PMID: 27597765 PMCID: PMC5027343 DOI: 10.14814/phy2.12902] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 07/27/2016] [Indexed: 11/24/2022] Open
Abstract
This study investigated miR‐148b as a potential physiological actor of physical inactivity‐induced effects in skeletal muscle. By using animal and human protocols, we demonstrated that the early phase of transition toward inactivity was associated with an increase in muscle miR‐148b content, which triggered the downregulation of NRAS and ROCK1 target genes. Using human myotubes, we demonstrated that overexpression of miR‐148b decreased NRAS and ROCK1 protein levels, and PKB phosphorylation and glucose uptake in response to insulin. Increase in muscle miR‐148b content might thus participate in the decrease in insulin sensitivity at the whole body level during the transition toward physical inactivity.
Collapse
Affiliation(s)
- Caroline Gastebois
- CarMeN Laboratory, INSERM U1060 INRA 1397 University of Lyon 1, Oullins, France
| | - Stéphanie Chanon
- CarMeN Laboratory, INSERM U1060 INRA 1397 University of Lyon 1, Oullins, France
| | - Sophie Rome
- CarMeN Laboratory, INSERM U1060 INRA 1397 University of Lyon 1, Oullins, France
| | - Christine Durand
- CarMeN Laboratory, INSERM U1060 INRA 1397 University of Lyon 1, Oullins, France
| | - Elise Pelascini
- Department of Digestive and Bariatric Surgery, Hospices Civils de Lyon, Lyon, France
| | - Audrey Jalabert
- CarMeN Laboratory, INSERM U1060 INRA 1397 University of Lyon 1, Oullins, France
| | - Vanessa Euthine
- CarMeN Laboratory, INSERM U1060 INRA 1397 University of Lyon 1, Oullins, France
| | | | - Stéphane Blanc
- Institut Pluridisciplinaire Hubert Curien, CNRS UMR 7178 University of Strasbourg, Strasbourg, France
| | - Chantal Simon
- CarMeN Laboratory, INSERM U1060 INRA 1397 University of Lyon 1, Oullins, France
| | - Etienne Lefai
- CarMeN Laboratory, INSERM U1060 INRA 1397 University of Lyon 1, Oullins, France
| |
Collapse
|
43
|
Regulation of hepatic Na+/K+-ATPase in obese female and male rats: involvement of ERK1/2, AMPK, and Rho/ROCK. Mol Cell Biochem 2017; 440:77-88. [DOI: 10.1007/s11010-017-3157-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 08/05/2017] [Indexed: 12/22/2022]
|
44
|
Amaya CN, Mitchell DC, Bryan BA. Rho kinase proteins display aberrant upregulation in vascular tumors and contribute to vascular tumor growth. BMC Cancer 2017; 17:485. [PMID: 28709411 PMCID: PMC5513090 DOI: 10.1186/s12885-017-3470-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 07/02/2017] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND The serine/threonine protein kinases ROCK1 and 2 are key RhoA-mediated regulators of cell shape and cytoskeletal dynamics. These proteins perform multiple functions in vascular endothelial cell physiology and are attractive targets for cancer therapy based on their roles as oncogenes and metastatic promoters. Given their critical functions in both of these processes, we hypothesized that molecular targeting of ROCK proteins would be exceedingly effective against vascular tumors such as hemangiomas and angiosarcomas, which are neoplasms composed of aberrant endothelial cells. METHODS In this study, we compared ROCK1 and 2 protein expression in a large panel of benign and malignant vascular tumors to that of normal vasculature. We then utilized shRNA technology to knockdown the expression of ROCK1 and 2 in SVR tumor-forming vascular cells, and evaluated tumor size and proliferation rate in a xenograft model. Finally, we employed proteomics and metabolomics to assess how knockdown of the ROCK paralogs induced alterations in protein expression/phosphorylation and metabolite concentrations in the xenograft tumors. RESULTS Our findings revealed that ROCK1 was overexpressed in malignant vascular tumors such as hemangioendotheliomas and angiosarcomas, and ROCK2 was overexpressed in both benign and malignant vascular tumors including hemangiomas, hemangioendotheliomas, hemangiopericytomas, and angiosarcomas. shRNA-mediated knockdown of ROCK2, but not ROCK1, in xenograft vascular tumors significantly reduced tumor size and proliferative index compared to control tumors. Proteomics and metabolomics analysis of the xenograft tumors revealed both overlapping as well as unique roles for the ROCK paralogs in regulating signal transduction and metabolite concentrations. CONCLUSIONS Collectively, these data indicate that ROCK proteins are overexpressed in diverse vascular tumors and suggest that specific targeting of ROCK2 proteins may show efficacy against malignant vascular tumors.
Collapse
Affiliation(s)
- Clarissa N Amaya
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, Center of Excellence in Cancer Research, 5001 El Paso Drive, MSB1 Room 2111, El Paso, TX, 79905, USA
| | - Dianne C Mitchell
- Minerva Genetics, 5130 Gateway Blvd East, Suite 315, El Paso, TX, 79905, USA
| | - Brad A Bryan
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, Center of Excellence in Cancer Research, 5001 El Paso Drive, MSB1 Room 2111, El Paso, TX, 79905, USA. .,Minerva Genetics, 5130 Gateway Blvd East, Suite 315, El Paso, TX, 79905, USA.
| |
Collapse
|
45
|
Functions of Rho family of small GTPases and Rho-associated coiled-coil kinases in bone cells during differentiation and mineralization. Biochim Biophys Acta Gen Subj 2017; 1861:1009-1023. [PMID: 28188861 DOI: 10.1016/j.bbagen.2017.02.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 02/02/2017] [Accepted: 02/06/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Members of Rho-associated coiled-coil kinases (ROCKs) are effectors of Rho family of small GTPases. ROCKs have multiple functions that include regulation of cellular contraction and polarity, adhesion, motility, proliferation, apoptosis, differentiation, maturation and remodeling of the extracellular matrix (ECM). SCOPE OF THE REVIEW Here, we focus on the action of RhoA and RhoA effectors, ROCK1 and ROCK2, in cells related to tissue mineralization: mesenchymal stem cells, chondrocytes, preosteoblasts, osteoblasts, osteocytes, lining cells and osteoclasts. MAJOR CONCLUSIONS The activation of the RhoA/ROCK pathway promotes stress fiber formation and reduces chondrocyte and osteogenic differentiations, in contrast to that in mesenchymal stem cells which stimulated the osteogenic and the chondrogenic differentiation. The effects of Rac1 and Cdc42 in promoting chondrocyte hypertrophy and of Rac1, Rac2 and Cdc42 in osteoclast are discussed. In addition, members of the Rho family of GTPases such Rac1, Rac2, Rac3 and Cdc42, acting upstream of ROCK and/or other protein effectors, may compensate the actions of RhoA, affecting directly or indirectly the actions of ROCKs as well as other protein effectors. GENERAL SIGNIFICANCE ROCK activity can trigger cartilage degradation and affect bone formation, therefore these kinases may represent a possible therapeutic target to treat osteoarthritis and osseous diseases. Inhibition of Rho/ROCK activity in chondrocytes prevents cartilage degradation, stimulate mineralization of osteoblasts and facilitate bone formation around implanted metals. Treatment with osteoprotegerin results in a significant decrease in the expression of Rho GTPases, ROCK1 and ROCK2, reducing bone resorption. Inhibition of ROCK signaling increases osteoblast differentiation in a topography-dependent manner.
Collapse
|
46
|
Chen J, Zhang J, Yang J, Xu L, Hu Q, Xu C, Yang S, Jiang H. Histone demethylase KDM3a, a novel regulator of vascular smooth muscle cells, controls vascular neointimal hyperplasia in diabetic rats. Atherosclerosis 2016; 257:152-163. [PMID: 28135625 DOI: 10.1016/j.atherosclerosis.2016.12.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 11/23/2016] [Accepted: 12/08/2016] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND AIMS Deregulation of histone demethylase KDM3a, an important regulator for H3K9 methylation, is correlated with obesity and abnormal metabolism in rodent models. However, the function of KDM3a in vascular remodeling under diabetic condition is unknown. METHODS Adenoviruses expressing KDM3a and lentiviruses expressing KDM3a-targeting siRNA were generated to study the role of KDM3a both in vivo and in vitro. The carotid artery balloon injury model was established in diabetic SD rats to evaluate the significance of KDM3a in vascular injury. RESULTS Diabetic vessels were associated with sustained loss of histone H3 lysine 9 di-methylation (H3K9me2) and elevation of KDM3a. This phenomenon was induced by high glucose (HG) and was persistently present even after removal from diabetic condition and high glucose in vascular smooth muscle cells (VSMCs). After 28-day balloon injury, KDM3a overexpression accelerated while KDM3a knockdown reduced neointima formation, following vascular injury in diabetic rats without glucose control. Microarray analysis revealed KDM3a altered the expression of vascular remodeling genes; particularly, it mediated the Rho/ROCK and AngII/AGTR1 pathways. In the in vivo study, HG and Ang II-stimulated proliferation and migration of VSMCs were enhanced by KDM3a overexpression, whereas markedly prevented by KDM3a knockdown. KDM3a regulated the transcription of AGTR1 and ROCK2 via controlling H3K9me2 in the proximal promoter regions. CONCLUSIONS Histone demethylase KDM3a promotes vascular neointimal hyperplasia in diabetic rats via AGTR1 and ROCK2 signaling pathways. Targeting KDM3a might represent a promising therapeutic approach for the prevention of coronary artery disease with diabetes.
Collapse
Affiliation(s)
- Jing Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jing Zhang
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, China
| | - Jian Yang
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, China
| | - Lin Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qi Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Changwu Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shuo Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
47
|
Rendina-Ruedy E, Graef J, Lightfoot S, Ritchey J, Clarke S, Lucas E, Smith B. Impaired glucose tolerance attenuates bone accrual by promoting the maturation of osteoblasts: Role of Beclin1-mediated autophagy. Bone Rep 2016; 5:199-207. [PMID: 28580387 PMCID: PMC5440954 DOI: 10.1016/j.bonr.2016.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 08/07/2016] [Indexed: 11/22/2022] Open
Abstract
Patients with type 2 diabetes mellitus (T2DM) experience a 1.5-3.5 fold increase in fracture risk, but the mechanisms responsible for these alterations in bone biomechanical properties remain elusive. Macroautophagy, often referred to as autophagy, is regulated by signaling downstream of the insulin receptor. Metabolic changes associated with the progression of glucose intolerance have been shown to alter autophagy in various tissues, but limited information is available in relation to bone cells. The aim of this study was to (a) investigate whether autophagy is altered in bone tissue during impaired glucose tolerance, and (b) determine how autophagy impacts osteoblast differentiation, activity, and maturation. Four-week-old, male C57BL/6 mice were fed a control (Con) or high fat (HF) diet for 2, 8, or 16 wks. Mice on the HF diet demonstrated elevated fasting blood glucose and impaired glucose tolerance. Reduced trabecular bone in the femoral neck was evident in the mice on the HF diet by 8 wks compared to Con mice. Histological evaluation of the tibia suggested that the high fat diet promoted terminal differentiation of the osteoblast to an osteocyte. This shift of the osteoblasts towards a non-mineralizing, osteocyte phenotype appears to be coordinated by Beclin1-mediated autophagy. Consistent with these changes in the osteoblast in vivo, the induction of autophagy was able to direct MC3T3-E1 cells towards a more mature osteoblast phenotype. Although these data are somewhat observational, further investigation is warranted to determine if Beclin1-mediated autophagy is essential for the terminal differentiation of the osteoblasts and whether autophagy is having a protective or deleterious effect on bone in T2DM.
Collapse
Key Words
- AGEs, advanced glycation end products
- AIN, American Institute of Nutrition
- AMPK, adenosine monophosphate-activated protein kinase
- Ambra1, vacuole sorting protein (Vps34/15), activating molecule in Beclin-1 regulator autophagy
- Atg, autophagy-related proteins
- BafA1, bafilomycinA1
- Beclin1, Bcl-2-interacting myosin-like coiled-coil protein
- FIP200, focal adhesion
- Hyperglycemia
- IR, insulin receptor
- Insulin
- LC3, microtubule associated light chain
- Macroautophagy
- Osteocyte
- PE, phosphatidylethanolamine
- ROCK1, rho kinase 1
- Rap, rapamycin
- T2DM, type 2 diabetes mellitus
- ULK1/2, unc-like kinase
- UVRAG, ultraviolet radiation resistance-associated gene
- mTORC1, mammalian or mechanistic target of rapamycin complex
Collapse
Affiliation(s)
- E. Rendina-Ruedy
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK, United States
| | - J.L. Graef
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK, United States
| | - S.A. Lightfoot
- Center for Cancer Prevention and Drug Development, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - J.W. Ritchey
- Department of Veterinary Pathobiology, Oklahoma State University, Stillwater, OK, United States
| | - S.L. Clarke
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK, United States
| | - E.A. Lucas
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK, United States
| | - B.J. Smith
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK, United States
| |
Collapse
|
48
|
Samuel MS, Rath N, Masre SF, Boyle ST, Greenhalgh DA, Kochetkova M, Bryson S, Stevenson D, Olson MF. Tissue-selective expression of a conditionally-active ROCK2-estrogen receptor fusion protein. Genesis 2016; 54:636-646. [PMID: 27775859 DOI: 10.1002/dvg.22988] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 10/12/2016] [Accepted: 10/20/2016] [Indexed: 11/10/2022]
Abstract
The serine/threonine kinases ROCK1 and ROCK2 are central mediators of actomyosin contractile force generation that act downstream of the RhoA small GTP-binding protein. As a result, they have key roles in regulating cell morphology and proliferation, and have been implicated in numerous pathological conditions and diseases including hypertension and cancer. Here we describe the generation of a gene-targeted mouse line that enables CRE-inducible expression of a conditionally-active fusion between the ROCK2 kinase domain and the hormone-binding domain of a mutated estrogen receptor (ROCK2:ER). This two-stage system of regulation allows for tissue-selective expression of the ROCK2:ER fusion protein, which then requires administration of estrogen analogues such as tamoxifen or 4-hydroxytamoxifen to elicit kinase activity. This conditional gain-of-function system was validated in multiple tissues by crossing with mice expressing CRE recombinase under the transcriptional control of cytokeratin14 (K14), murine mammary tumor virus (MMTV) or cytochrome P450 Cyp1A1 (Ah) promoters, driving appropriate expression in the epidermis, mammary or intestinal epithelia respectively. Given the interest in ROCK signaling in normal physiology and disease, this mouse line will facilitate research into the consequences of ROCK activation that could be used to complement conditional knockout models. Birth Defects Research (Part A) 106:636-646, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Michael S Samuel
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Frome Road, Adelaide, South Australia, 5000, Australia
- School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, South Australia, 5000, Australia
| | - Nicola Rath
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Siti F Masre
- Biomedical Science Programme, School of Diagnostic and Applied Health Sciences, Faculty of Allied Health Sciences, University of Kebangsaan, Kuala Lumpur, 50300, Malaysia
- Section of Dermatology and Molecular Carcinogenesis College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Sarah T Boyle
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Frome Road, Adelaide, South Australia, 5000, Australia
| | - David A Greenhalgh
- Section of Dermatology and Molecular Carcinogenesis College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Marina Kochetkova
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Frome Road, Adelaide, South Australia, 5000, Australia
| | - Sheila Bryson
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - David Stevenson
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Michael F Olson
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| |
Collapse
|
49
|
Nandipati KC, Subramanian S, Agrawal DK. Protein kinases: mechanisms and downstream targets in inflammation-mediated obesity and insulin resistance. Mol Cell Biochem 2016; 426:27-45. [PMID: 27868170 DOI: 10.1007/s11010-016-2878-8] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 11/07/2016] [Indexed: 12/23/2022]
Abstract
Obesity-induced low-grade inflammation (metaflammation) impairs insulin receptor signaling. This has been implicated in the development of insulin resistance. Insulin signaling in the target tissues is mediated by stress kinases such as p38 mitogen-activated protein kinase, c-Jun NH2-terminal kinase, inhibitor of NF-kB kinase complex β (IKKβ), AMP-activated protein kinase, protein kinase C, Rho-associated coiled-coil containing protein kinase, and RNA-activated protein kinase. Most of these kinases phosphorylate several key regulators in glucose homeostasis. The phosphorylation of serine residues in the insulin receptor and IRS-1 molecule results in diminished enzymatic activity in the phosphatidylinositol 3-kinase (PI3K)/Akt pathway. This has been one of the key mechanisms observed in the tissues that are implicated in insulin resistance especially in type 2 diabetes mellitus (T2-DM). Identifying the specific protein kinases involved in obesity-induced chronic inflammation may help in developing the targeted drug therapies to minimize the insulin resistance. This review is focused on the protein kinases involved in the inflammatory cascade and molecular mechanisms and their downstream targets with special reference to obesity-induced T2-DM.
Collapse
Affiliation(s)
- Kalyana C Nandipati
- Department of Surgery, Creighton University School of Medicine, 601 N. 30th Street, Suite # 3700, Omaha, NE, 68131, USA.
- Department of Clinical & Translational Science, Creighton University School of Medicine, 2500, California Plaza, Room # 510, Criss II, Omaha, NE, 68131, USA.
| | - Saravanan Subramanian
- Department of Clinical & Translational Science, Creighton University School of Medicine, 2500, California Plaza, Room # 510, Criss II, Omaha, NE, 68131, USA
| | - Devendra K Agrawal
- Department of Clinical & Translational Science, Creighton University School of Medicine, 2500, California Plaza, Room # 510, Criss II, Omaha, NE, 68131, USA
| |
Collapse
|
50
|
Sousa-Lima I, Park SY, Chung M, Jung HJ, Kang MC, Gaspar JM, Seo JA, Macedo MP, Park KS, Mantzoros C, Lee SH, Kim YB. Methylsulfonylmethane (MSM), an organosulfur compound, is effective against obesity-induced metabolic disorders in mice. Metabolism 2016; 65:1508-21. [PMID: 27621186 DOI: 10.1016/j.metabol.2016.07.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 07/06/2016] [Accepted: 07/14/2016] [Indexed: 01/20/2023]
Abstract
Methylsulfonylmethane (MSM), an organosulfur compound, has been used as a dietary supplement that can improve various metabolic diseases. However, the effect of MSM on obesity-linked metabolic disorders remains unclear. The goal of the current study is to determine whether MSM has beneficial effects on glucose and lipid homeostasis in obesity-associated pathophysiologic states. High-fat diet-induced obese (DIO) and genetically obese diabetic db/db mice treated with MSM (1%-5% v/v, by drinking water) were studied. Metabolic parameters involved in glucose and lipid metabolism were determined. Treatment of DIO mice with MSM leads to a significant decrease in blood glucose levels. DIO mice treated with MSM are hypersensitive to insulin, as evidenced by decreased serum insulin and an increase in the area above the curve during an ITT. Concurrently, MSM reduces hepatic triglyceride and cholesterol contents in DIO mice. These effects are accompanied by reductions in gene expression of key molecules involved in lipogenesis and inflammation. FACS analysis reveals that MSM markedly increases the frequency of B cells and decreases the frequency of myeloid cells in peripheral blood and in bone marrow. Moreover, overnutrition-induced changes of femur microarchitecture are restored by MSM. In db/db mice, a marked impairment in glucose and lipid metabolic profiles is notably ameliorated when MSM is supplemented. These data suggest that MSM has beneficial effects on multiple metabolic dysfunctions, including hyperglycemia, hyperinsulinemia, insulin resistance, and inflammation. Thus, MSM could be the therapeutic option for the treatment of obesity-related metabolic disorders such as type 2 diabetes and fatty liver diseases.
Collapse
Affiliation(s)
- Inês Sousa-Lima
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA; Centro de Estudos de Doenças Crónicas CEDOC, NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Shin-Young Park
- Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Michelle Chung
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Hyun Ju Jung
- Department of Oriental Pharmacy, Wonkwang University, Iksan, Jeonbuk, Republic of Korea
| | - Min-Cheol Kang
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Joana M Gaspar
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA; Centro de Estudos de Doenças Crónicas CEDOC, NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Ji A Seo
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - M Paula Macedo
- Centro de Estudos de Doenças Crónicas CEDOC, NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal; APDP - Education and Research Center, Lisbon, Portugal
| | - Kyong Soo Park
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University; Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Christos Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | | | - Young-Bum Kim
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA; Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University.
| |
Collapse
|