1
|
Varshini MS, Reddy RA, Krishnamurthy PT, Wadhwani A. Harmony of Wnt pathway in Alzheimer's: Navigating the multidimensional progression from preclinical to clinical stages. Neurosci Biobehav Rev 2024; 165:105863. [PMID: 39179059 DOI: 10.1016/j.neubiorev.2024.105863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 08/14/2024] [Accepted: 08/18/2024] [Indexed: 08/26/2024]
Abstract
The Wnt pathway stands out as a pivotal signal transduction pathway, operating through two distinct modes of signaling: the canonical/β-catenin pathway and the non-canonical pathway. Among these, the canonical pathway assumes a paramount role in various physiological and pathological processes within the human body. Particularly in the brain, Wnt exhibits involvement in fundamental physiological events including neuronal differentiation/survival, axonogenesis, neural stem cell regulation, synaptic plasticity, and cell cycle modulation. Notably, scientific evidence underscores the critical role of the Wnt pathway in the pathogenesis of Alzheimer's disease (AD), correlating with its involvement in key pathological features such as tau tangles, Amyloid-β plaques, synaptic dysfunction, oxidative stress, mitochondrial dysfunction, cognitive impairments, and disruption of the blood-brain barrier integrity. This review aims to comprehensively explore the involvement and significance of Wnt signaling in Alzheimer's. Furthermore, it delves into recent advancements in research on Wnt signaling, spanning from preclinical investigations to clinical trials.
Collapse
Affiliation(s)
- Magham Sai Varshini
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, TN 643001, India
| | - Ramakkamma Aishwarya Reddy
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, TN 643001, India
| | | | - Ashish Wadhwani
- Department of Pharmaceutical Biotechnology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, TN 643001, India; Faculty of Health Sciences, School of Pharmacy, JSS Academy of Higher Education and Research, Mauritius, Vacoas 73304, Mauritius
| |
Collapse
|
2
|
Sai Varshini M, Aishwarya Reddy R, Thaggikuppe Krishnamurthy P. Unlocking hope: GSK-3 inhibitors and Wnt pathway activation in Alzheimer's therapy. J Drug Target 2024; 32:909-917. [PMID: 38838023 DOI: 10.1080/1061186x.2024.2365263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/07/2024]
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder characterised by progressive cognitive decline and the accumulation of amyloid-β plaques and tau tangles. The Wnt signalling pathway known for its crucial role in neurodevelopment and adult neurogenesis has emerged as a potential target for therapeutic intervention in AD. Glycogen synthase kinase-3 beta (GSK-3β), a key regulator of the Wnt pathway, plays a pivotal role in AD pathogenesis by promoting tau hyperphosphorylation and neuroinflammation. Several preclinical studies have demonstrated that inhibiting GSK-3β leads to the activation of Wnt pathway thereby promoting neuroprotective effects, and mitigating cognitive deficits in AD animal models. The modulation of Wnt signalling appears to have multifaceted benefits including the reduction of amyloid-β production, tau hyperphosphorylation, enhancement of synaptic plasticity, and inhibition of neuroinflammation. These findings suggest that targeting GSK-3β to activate Wnt pathway may represent a novel approach for slowing or halting the progression of AD. This hypothesis reviews the current state of research exploring the activation of Wnt pathway through the inhibition of GSK-3β as a promising therapeutic strategy in AD.
Collapse
Affiliation(s)
- Magham Sai Varshini
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, India
| | - Ramakkamma Aishwarya Reddy
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, India
| | | |
Collapse
|
3
|
Umbarkar P, Ejantkar S, Ruiz Ramirez SY, Toro Cora A, Zhang Q, Tousif S, Lal H. Cardiac fibroblast GSK-3α aggravates ischemic cardiac injury by promoting fibrosis, inflammation, and impairing angiogenesis. Basic Res Cardiol 2023; 118:35. [PMID: 37656238 PMCID: PMC11340261 DOI: 10.1007/s00395-023-01005-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/14/2023] [Accepted: 08/16/2023] [Indexed: 09/02/2023]
Abstract
Myocardial infarction (MI) is the leading cause of death worldwide. Glycogen synthase kinase-3 (GSK-3) has been considered to be a promising therapeutic target for cardiovascular diseases. GSK-3 is a family of ubiquitously expressed serine/threonine kinases. GSK-3 isoforms appear to play overlapping, unique, and even opposing functions in the heart. Previously, our group identified that cardiac fibroblast (FB) GSK-3β acts as a negative regulator of fibrotic remodeling in the ischemic heart. However, the role of FB-GSK-3α in MI pathology is not defined. To determine the role of FB-GSK-3α in MI-induced adverse cardiac remodeling, GSK-3α was deleted specifically in the residential fibroblast or myofibroblast (MyoFB) using tamoxifen (TAM) inducible Tcf21 or Periostin (Postn) promoter-driven Cre recombinase, respectively. Echocardiographic analysis revealed that FB- or MyoFB-specific GSK-3α deletion prevented the development of dilative remodeling and cardiac dysfunction. Morphometrics and histology studies confirmed improvement in capillary density and a remarkable reduction in hypertrophy and fibrosis in the KO group. We harvested the hearts at 4 weeks post-MI and analyzed signature genes of adverse remodeling. Specifically, qPCR analysis was performed to examine the gene panels of inflammation (TNFα, IL-6, IL-1β), fibrosis (COL1A1, COL3A1, COMP, Fibronectin-1, Latent TGF-β binding protein 2), and hypertrophy (ANP, BNP, MYH7). These molecular markers were essentially normalized due to FB-specific GSK-3α deletion. Further molecular studies confirmed that FB-GSK-3α could regulate NF-kB activation and expression of angiogenesis-related proteins. Our findings suggest that FB-GSK-3α plays a critical role in the pathological cardiac remodeling of ischemic hearts, therefore, it could be therapeutically targeted.
Collapse
Affiliation(s)
- Prachi Umbarkar
- Division of Cardiovascular Disease, UAB|The University of Alabama at Birmingham, 1720 2nd Ave South, Birmingham, AL, 35294-1913, USA.
| | - Suma Ejantkar
- Division of Cardiovascular Disease, UAB|The University of Alabama at Birmingham, 1720 2nd Ave South, Birmingham, AL, 35294-1913, USA
| | - Sulivette Y Ruiz Ramirez
- Division of Cardiovascular Disease, UAB|The University of Alabama at Birmingham, 1720 2nd Ave South, Birmingham, AL, 35294-1913, USA
| | - Angelica Toro Cora
- Division of Cardiovascular Disease, UAB|The University of Alabama at Birmingham, 1720 2nd Ave South, Birmingham, AL, 35294-1913, USA
| | - Qinkun Zhang
- Division of Cardiovascular Disease, UAB|The University of Alabama at Birmingham, 1720 2nd Ave South, Birmingham, AL, 35294-1913, USA
| | - Sultan Tousif
- Division of Cardiovascular Disease, UAB|The University of Alabama at Birmingham, 1720 2nd Ave South, Birmingham, AL, 35294-1913, USA
| | - Hind Lal
- Division of Cardiovascular Disease, UAB|The University of Alabama at Birmingham, 1720 2nd Ave South, Birmingham, AL, 35294-1913, USA.
| |
Collapse
|
4
|
Umbarkar P, Ruiz Ramirez SY, Toro Cora A, Tousif S, Lal H. GSK-3 at the heart of cardiometabolic diseases: Isoform-specific targeting is critical to therapeutic benefit. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166724. [PMID: 37094727 PMCID: PMC10247467 DOI: 10.1016/j.bbadis.2023.166724] [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: 01/23/2023] [Revised: 04/14/2023] [Accepted: 04/18/2023] [Indexed: 04/26/2023]
Abstract
Glycogen synthase kinase-3 (GSK-3) is a family of serine/threonine kinases. The GSK-3 family has 2 isoforms, GSK-3α and GSK-3β. The GSK-3 isoforms have been shown to play overlapping as well as isoform-specific-unique roles in both, organ homeostasis and the pathogenesis of multiple diseases. In the present review, we will particularly focus on expanding the isoform-specific role of GSK-3 in the pathophysiology of cardiometabolic disorders. We will highlight recent data from our lab that demonstrated the critical role of cardiac fibroblast (CF) GSK-3α in promoting injury-induced myofibroblast transformation, adverse fibrotic remodeling, and deterioration of cardiac function. We will also discuss studies that found the exact opposite role of CF-GSK-3β in cardiac fibrosis. We will review emerging studies with inducible cardiomyocyte (CM)-specific as well as global isoform-specific GSK-3 KOs that demonstrated inhibition of both GSK-3 isoforms provides benefits against obesity-associated cardiometabolic pathologies. The underlying molecular interactions and crosstalk among GSK-3 and other signaling pathways will be discussed. We will briefly review the specificity and limitations of the available small molecule inhibitors targeting GSK-3 and their potential applications to treat metabolic disorders. Finally, we will summarize these findings and offer our perspective on envisioning GSK-3 as a therapeutic target for the management of cardiometabolic diseases.
Collapse
Affiliation(s)
- Prachi Umbarkar
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Sulivette Y Ruiz Ramirez
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Angelica Toro Cora
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Sultan Tousif
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Hind Lal
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
5
|
Zhang GC, Wu YJ, Liu FQ, Chen Q, Sun XY, Qu QY, Fu HX, Huang XJ, Zhang XH. β2-adrenergic receptor agonist corrects immune thrombocytopenia by reestablishing the homeostasis of T cell differentiation. J Thromb Haemost 2023; 21:1920-1933. [PMID: 36972787 DOI: 10.1016/j.jtha.2023.02.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 02/22/2023] [Accepted: 02/23/2023] [Indexed: 03/29/2023]
Abstract
BACKGROUND The sympathetic nerve is known to regulate immune responses in autoimmunity. Aberrant T cell immunity plays a vital role in immune thrombocytopenia (ITP) pathogenesis. The spleen is the primary site of platelet destruction. However, little is known whether and how splenic sympathetic innervation and neuroimmune modulation contribute to ITP pathogenesis. OBJECTIVES To determine the sympathetic distribution in the spleen of ITP mice and the association between splenic sympathetic nerves and T cell immunity in ITP development, and to evaluate the treatment potential of β2-adrenergic receptor (β2-AR) in ITP. METHODS Chemical sympathectomy was performed in an ITP mouse model with 6-hydroxydopamine and treated with β2-AR agonists to evaluate the effects of sympathetic denervation and activation. RESULTS Decreased sympathetic innervation in the spleen of ITP mice was observed. Significantly increased percentages of Th1 and Tc1 cells and reduced percentages of regulatory T cells (Tregs) were also observed in ITP mice with chemical sympathectomy (ITP-syx mice) relative to mice without sympathectomy (controls). Expression of genes associated with Th1, including IFN-γ and IRF8, was significantly upregulated, whereas genes associated with Tregs, including Foxp3 and CTLA4, were significantly downregulated in ITP-syx mice compared with controls. Furthermore, β2-AR restored the percentage of Tregs and increased platelet counts at days 7 and 14 in ITP mice. CONCLUSION Our findings indicate that decreased sympathetic distribution contributes to ITP pathogenesis by disturbing the homeostasis of T cells and that β2-AR agonists have potential as a novel treatment for ITP.
Collapse
Affiliation(s)
- Gao-Chao Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Ye-Jun Wu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Feng-Qi Liu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Qi Chen
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Xue-Yan Sun
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Qing-Yuan Qu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Hai-Xia Fu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Xiao-Jun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Xiao-Hui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; National Clinical Research Center for Hematologic Disease, Beijing, China.
| |
Collapse
|
6
|
Wang W, Guo L, Jiang B, Yan B, Li Y, Ye X, Yang Y, Liu S, Shao Z, Diao H. Role of the Glycogen Synthase Kinase 3-Cyclic AMP/Protein Kinase A in the Immobilization of Human Sperm by Tideglusib. Reprod Sci 2023; 30:1281-1290. [PMID: 36207578 DOI: 10.1007/s43032-022-01086-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 09/15/2022] [Indexed: 10/10/2022]
Abstract
Tideglusib is considered to be a promising alternative to nonyl alcohol-9 contraceptives. Previous studies have demonstrated that the rapid spermicidal effect of tideglusib at a high concentration (≥ 10 μM) may occur through detergent-like activity; however, the effect of low concentrations of tideglusib (< 5 μM) on sperm is unknown. We explored the intracellular mechanism of tideglusib (< 5 μM) on the immobilization of human sperm by exploring related signaling pathways in human sperm. After treatment with tideglusib (1.25 μM) for 2 h, sperm motility rate decreased to 0, while sperm membrane integrity rate was 70%. Protein tyrosine phosphorylation level and intracellular cyclic adenosine 3,5-monophosphate (cAMP) concentration decreased significantly compared to those in the control group. Isobutylmethylxanthine and 8-Bromo-cAMP relieved the inhibition of spermatozoa tyrosine phosphorylation, while tyrosine phosphorylation of sperm protein in the H89 and CALP1 treatment groups was significantly inhibited, and there was no difference in the tideglusib treatment group. H-89 and CALP1 reduced the level of serine phosphorylation of GSK-3α/β (Ser21/9), while its level was enhanced by IBMX and 8-Bromo-cAMP. Our results show the existence of the GSK3-cAMP/PKA regulatory loop in human sperm, which may mediate the immobilization effect of tideglusib at low of concentrations (e.g., 1.25 μM) on sperm motility.
Collapse
Affiliation(s)
- Weiwei Wang
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Lina Guo
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Bingbing Jiang
- College of Chemistry and Chemical Engineering, Donghua University, Shanghai, 201620, China
| | - Bin Yan
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yuhua Li
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Xin Ye
- College of Chemistry and Chemical Engineering, Donghua University, Shanghai, 201620, China
| | - Yiting Yang
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Suying Liu
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhiyu Shao
- College of Chemistry and Chemical Engineering, Donghua University, Shanghai, 201620, China.
| | - Hua Diao
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
7
|
Tousif S, Singh AP, Umbarkar P, Galindo C, Wheeler N, Coro AT, Zhang Q, Prabhu SD, Lal H. Ponatinib Drives Cardiotoxicity by S100A8/A9-NLRP3-IL-1β Mediated Inflammation. Circ Res 2023; 132:267-289. [PMID: 36625265 PMCID: PMC9898181 DOI: 10.1161/circresaha.122.321504] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 12/28/2022] [Indexed: 01/11/2023]
Abstract
BACKGROUND The tyrosine kinase inhibitor ponatinib is the only treatment option for chronic myelogenous leukemia patients with T315I (gatekeeper) mutation. Pharmacovigilance analysis of Food and Drug Administration and World Health Organization datasets has revealed that ponatinib is the most cardiotoxic agent among all Food and Drug Administration-approved tyrosine kinase inhibitors in a real-world scenario. However, the mechanism of ponatinib-induced cardiotoxicity is unknown. METHODS The lack of well-optimized mouse models has hampered the in vivo cardio-oncology studies. Here, we show that cardiovascular comorbidity mouse models evidence a robust cardiac pathological phenotype upon ponatinib treatment. A combination of multiple in vitro and in vivo models was employed to delineate the underlying molecular mechanisms. RESULTS An unbiased RNA sequencing analysis identified the enrichment of dysregulated inflammatory genes, including a multifold upregulation of alarmins S100A8/A9, as a top hit in ponatinib-treated hearts. Mechanistically, we demonstrate that ponatinib activates the S100A8/A9-TLR4 (Toll-like receptor 4)-NLRP3 (NLR family pyrin domain-containing 3)-IL (interleukin)-1β signaling pathway in cardiac and systemic myeloid cells, in vitro and in vivo, thereby leading to excessive myocardial and systemic inflammation. Excessive inflammation was central to the cardiac pathology because interventions with broad-spectrum immunosuppressive glucocorticoid dexamethasone or specific inhibitors of NLRP3 (CY-09) or S100A9 (paquinimod) nearly abolished the ponatinib-induced cardiac dysfunction. CONCLUSIONS Taken together, these findings uncover a novel mechanism of ponatinib-induced cardiac inflammation leading to cardiac dysfunction. From a translational perspective, our results provide critical preclinical data and rationale for a clinical investigation into immunosuppressive interventions for managing ponatinib-induced cardiotoxicity.
Collapse
Affiliation(s)
- Sultan Tousif
- Division of Cardiovascular Disease, UAB | The University of Alabama at Birmingham, Birmingham, AL
| | - Anand P. Singh
- Division of Cardiovascular Disease, UAB | The University of Alabama at Birmingham, Birmingham, AL
| | - Prachi Umbarkar
- Division of Cardiovascular Disease, UAB | The University of Alabama at Birmingham, Birmingham, AL
| | - Cristi Galindo
- Department of Biology, Western Kentucky University, Bowling Green, KY 42101, USA35294-1913, USA
| | - Nicholas Wheeler
- Department of Biology, Western Kentucky University, Bowling Green, KY 42101, USA35294-1913, USA
| | - Angelica Toro Coro
- Division of Cardiovascular Disease, UAB | The University of Alabama at Birmingham, Birmingham, AL
| | - Qinkun Zhang
- Division of Cardiovascular Disease, UAB | The University of Alabama at Birmingham, Birmingham, AL
| | - Sumanth D. Prabhu
- Division of Cardiology, Department of Medicine, Washington University in St. Louis
| | - Hind Lal
- Division of Cardiovascular Disease, UAB | The University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
8
|
Rousseau B, Murugan S, Palagani A, Sarkar DK. Beta 2 adrenergic receptor and mu opioid receptor interact to potentiate the aggressiveness of human breast cancer cell by activating the glycogen synthase kinase 3 signaling. Breast Cancer Res 2022; 24:33. [PMID: 35568869 PMCID: PMC9107672 DOI: 10.1186/s13058-022-01526-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 04/20/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Opioid and beta-adrenergic receptors are recently shown to cross talk via formation of receptor heterodimers to control the growth and proliferation of breast cancer cells. However, the underlying cell signaling mechanism remained unclear. METHODS To determine the effect of the interaction of the two systems in breast cancer, we employed triple-negative breast cancer cell lines MDA-MB-231 and MDA-MB-468, CRISPR or chemical inhibition or activation of beta-adrenergic receptors (B2AR) and mu-opioid receptors (MOR) gene, and PCR array technology and studied aggressive tumor phenotype and signaling cascades. RESULTS We show here that in triple-negative breast cancer cells, the reduction in expression B2AR and MOR by genetic and pharmacological tools leads to a less aggressive phenotype of triple-negative breast cancer cells in vitro and in animal xenografts. Genomic analysis indicates the glycogen synthase kinase 3 (GSK3) pathway as a possible candidate messenger system involved in B2AR and MOR cross talk. GSK3 inactivation in MDA-MB-231 and MDA-MB-468 cells induced similar phenotypic changes as the inhibition of B2AR and/or MOR, while a GSK3 activation by wortmannin reversed the effects of B2AR and/or MOR knockdown on these cells. GSK3 inactivation also prevents B2AR agonist norepinephrine or MOR agonist DAMGO from affecting MDA-MB-231 and MDA-MB-468 cell proliferation. CONCLUSIONS These data confirm a role of B2AR and MOR interaction in the control of breast cancer cell growth and identify a possible role of the GSK3 signaling system in mediation of these two receptors' cross talk. Screening for ligands targeting B2AR and MOR interaction and/or the GSK3 system may help to identify novel drugs for the prevention of triple-negative breast cancer cell growth and metastasis.
Collapse
Affiliation(s)
- Bénédicte Rousseau
- Endocrine Research Program, Department of Animal Sciences, Rutgers, The State University of New Jersey, 67 Poultry Farm Lane, New Brunswick, NJ, 08901, USA
| | - Sengottuvelan Murugan
- Endocrine Research Program, Department of Animal Sciences, Rutgers, The State University of New Jersey, 67 Poultry Farm Lane, New Brunswick, NJ, 08901, USA
| | - Ajay Palagani
- Endocrine Research Program, Department of Animal Sciences, Rutgers, The State University of New Jersey, 67 Poultry Farm Lane, New Brunswick, NJ, 08901, USA
| | - Dipak K Sarkar
- Endocrine Research Program, Department of Animal Sciences, Rutgers, The State University of New Jersey, 67 Poultry Farm Lane, New Brunswick, NJ, 08901, USA.
| |
Collapse
|
9
|
Cao X, Wu W, Wang D, Sun W, Lai S. Glycogen synthase kinase GSK3α promotes tumorigenesis by activating HIF1/VEGFA signaling pathway in NSCLC tumor. Cell Commun Signal 2022; 20:32. [PMID: 35292059 PMCID: PMC8922767 DOI: 10.1186/s12964-022-00825-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/06/2022] [Indexed: 12/24/2022] Open
Abstract
Background Lung cancer is one of the most common cancers and the leading cause of cancer-related death. Glycogen synthase kinase-3 (GSK-3) α, a member of the glycogen synthase kinase-3 family, reportedly plays a role in tumorigenesis. However, its biological function in tumorigenesis requires deeper exploration. Hypoxia is a major feature of solid tumor, along with decreasing availability of oxygen, inducing treatment resistance, and tumor progress. Methods Levels of GSK3α expression in clinical samples were detected using western blot and IHC assays, while its biological function and underlying mechanism of action in tumor progression were investigated using western blot, CCK8, cell cycle, colony formation, Transwell, ELISA and tube formation assays. Furthermore, we investigated the relationship between GSK3α expression and the HIF1α/VEGFA signaling pathway in vivo using a mouse xenograft model. Results GSK3α was significantly upregulated in NSCLC patients with cases that exhibited high GSK3α levels recording shorter survival times. Moreover, GSK3α overexpression promoted proliferation, migration, invasion and clone formation ability of NSCLC cells, while its silencing resulted in an opposite phenomenon. Moreover, GSK3α not only activated the HIF1α/VEGFA signaling pathway, but also regulated HIF1α stabilization independently via the PHDs-pVHL signaling pathway. Moreover, GSK3α-mediated tumor angiogenesis depended on HIF1α expression both in vitro and in vivo. Conclusion GSK3α functioned as an oncogene in NSCLC tumorigenesis by regulating the HIF1/VEGFA signaling pathway in an independent manner through the PHDs-pVHL signaling pathway. These findings were expected to provide novel sights to guide future development of therapies for effective treatment of NSCLC. Video abstract
Supplementary Information The online version contains supplementary material available at 10.1186/s12964-022-00825-3.
Collapse
Affiliation(s)
- Xiaonian Cao
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wei Wu
- Department of Hepatopancreatobiliary Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Gusao Tree Road No. 16 of Jianghan District, Wuhan, 430000, Hubei Province, China
| | - Dao Wang
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wei Sun
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Senyan Lai
- Department of Gastrointestinal Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
10
|
Gupte M, Tousif S, Lemon JJ, Toro Cora A, Umbarkar P, Lal H. Isoform-Specific Role of GSK-3 in High Fat Diet Induced Obesity and Glucose Intolerance. Cells 2022; 11:cells11030559. [PMID: 35159367 PMCID: PMC8834358 DOI: 10.3390/cells11030559] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/26/2022] [Accepted: 02/01/2022] [Indexed: 02/07/2023] Open
Abstract
Obesity-associated metabolic disorders are rising to pandemic proportions; hence, there is an urgent need to identify underlying molecular mechanisms. Glycogen synthase kinase-3 (GSK-3) signaling is highly implicated in metabolic diseases. Furthermore, GSK-3 expression and activity are increased in Type 2 diabetes patients. However, the isoform-specific role of GSK-3 in obesity and glucose intolerance is unclear. Pharmacological GSK-3 inhibitors are not isoform-specific, and tissue-specific genetic models are of limited value to predict the clinical outcome of systemic inhibiion. To overcome these limitations, we created novel mouse models of ROSA26CreERT2-driven, tamoxifen-inducible conditional deletion of GSK-3 that allowed us to delete the gene globally in an isoform-specific and temporal manner. Isoform-specific GSK-3 KOs and littermate controls were subjected to a 16-week high-fat diet (HFD) protocol. On an HFD, GSK-3α KO mice had a significantly lower body weight and modest improvement in glucose tolerance compared to their littermate controls. In contrast, GSK-3β-deletion-mediated improved glucose tolerance was evident much earlier in the timeline and extended up to 12 weeks post-HFD. However, this protective effect weakened after chronic HFD (16 weeks) when GSK-3β KO mice had a significantly higher body weight compared to controls. Importantly, GSK-3β KO mice on a control diet maintained significant improvement in glucose tolerance even after 16 weeks. In summary, our novel mouse models allowed us to delineate the isoform-specific role of GSK-3 in obesity and glucose tolerance. From a translational perspective, our findings underscore the importance of maintaining a healthy weight in patients receiving lithium therapy, which is thought to work by GSK-3 inhibition mechanisms.
Collapse
Affiliation(s)
- Manisha Gupte
- Department of Biology, Austin Peay State University, Clarksville, TN 37044, USA;
- Correspondence: (M.G.); (H.L.)
| | - Sultan Tousif
- Division of Cardiovascular Diseases, The University of Alabama at Birmingham (UAB), Birmingham, AL 35233, USA; (S.T.); (A.T.C.); (P.U.)
| | - Jacob J. Lemon
- Department of Biology, Austin Peay State University, Clarksville, TN 37044, USA;
| | - Angelica Toro Cora
- Division of Cardiovascular Diseases, The University of Alabama at Birmingham (UAB), Birmingham, AL 35233, USA; (S.T.); (A.T.C.); (P.U.)
| | - Prachi Umbarkar
- Division of Cardiovascular Diseases, The University of Alabama at Birmingham (UAB), Birmingham, AL 35233, USA; (S.T.); (A.T.C.); (P.U.)
| | - Hind Lal
- Division of Cardiovascular Diseases, The University of Alabama at Birmingham (UAB), Birmingham, AL 35233, USA; (S.T.); (A.T.C.); (P.U.)
- Correspondence: (M.G.); (H.L.)
| |
Collapse
|
11
|
Wang L, Li J, Di LJ. Glycogen synthesis and beyond, a comprehensive review of GSK3 as a key regulator of metabolic pathways and a therapeutic target for treating metabolic diseases. Med Res Rev 2021; 42:946-982. [PMID: 34729791 PMCID: PMC9298385 DOI: 10.1002/med.21867] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 08/01/2021] [Accepted: 10/24/2021] [Indexed: 12/19/2022]
Abstract
Glycogen synthase kinase‐3 (GSK3) is a highly evolutionarily conserved serine/threonine protein kinase first identified as an enzyme that regulates glycogen synthase (GS) in response to insulin stimulation, which involves GSK3 regulation of glucose metabolism and energy homeostasis. Both isoforms of GSK3, GSK3α, and GSK3β, have been implicated in many biological and pathophysiological processes. The various functions of GSK3 are indicated by its widespread distribution in multiple cell types and tissues. The studies of GSK3 activity using animal models and the observed effects of GSK3‐specific inhibitors provide more insights into the roles of GSK3 in regulating energy metabolism and homeostasis. The cross‐talk between GSK3 and some important energy regulators and sensors and the regulation of GSK3 in mitochondrial activity and component function further highlight the molecular mechanisms in which GSK3 is involved to regulate the metabolic activity, beyond its classical regulatory effect on GS. In this review, we summarize the specific roles of GSK3 in energy metabolism regulation in tissues that are tightly associated with energy metabolism and the functions of GSK3 in the development of metabolic disorders. We also address the impacts of GSK3 on the regulation of mitochondrial function, activity and associated metabolic regulation. The application of GSK3 inhibitors in clinical tests will be highlighted too. Interactions between GSK3 and important energy regulators and GSK3‐mediated responses to different stresses that are related to metabolism are described to provide a brief overview of previously less‐appreciated biological functions of GSK3 in energy metabolism and associated diseases through its regulation of GS and other functions.
Collapse
Affiliation(s)
- Li Wang
- Proteomics, Metabolomics, and Drug Development Core, Faculty of Health Sciences, University of Macau, Macau, China.,Department of Biomedical Sciences, Faculty of Health Sciences, Macau, China.,Cancer Center of the Faculty of Health Sciences, University of Macau, Macau, China.,Institute of Translational Medicine, University of Macau, Macau, China.,Ministry of Education, Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Jiajia Li
- Department of Biomedical Sciences, Faculty of Health Sciences, Macau, China.,Cancer Center of the Faculty of Health Sciences, University of Macau, Macau, China.,Institute of Translational Medicine, University of Macau, Macau, China.,Ministry of Education, Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Li-Jun Di
- Department of Biomedical Sciences, Faculty of Health Sciences, Macau, China.,Cancer Center of the Faculty of Health Sciences, University of Macau, Macau, China.,Institute of Translational Medicine, University of Macau, Macau, China.,Ministry of Education, Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| |
Collapse
|
12
|
Hall EJ, Pal S, Glennon MS, Shridhar P, Satterfield SL, Weber B, Zhang Q, Salama G, Lal H, Becker JR. Cardiac natriuretic peptide deficiency sensitizes the heart to stress induced ventricular arrhythmias via impaired CREB signaling. Cardiovasc Res 2021; 118:2124-2138. [PMID: 34329394 DOI: 10.1093/cvr/cvab257] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/28/2021] [Indexed: 11/14/2022] Open
Abstract
AIMS The cardiac natriuretic peptides (atrial natriuretic peptide [ANP] and B-type natriuretic peptide [BNP]) are important regulators of cardiovascular physiology, with reduced natriuretic peptide (NP) activity linked to multiple human cardiovascular diseases. We hypothesized that deficiency of either ANP or BNP would lead to similar changes in left ventricular structure and function given their shared receptor affinities. METHODS AND RESULTS We directly compared murine models deficient of ANP or BNP in the same genetic backgrounds (C57BL6/J) and environments. We evaluated control, ANP deficient (Nppa-/-) or BNP deficient (Nppb-/-) mice under unstressed conditions and multiple forms of pathological myocardial stress. Survival, myocardial structure, function and electrophysiology, tissue histology, and biochemical analyses were evaluated in the groups. In vitro validation of our findings was performed using human derived induced pluripotent stem cell cardiomyocytes (iPS-CM). In the unstressed state, both ANP and BNP deficient mice displayed mild ventricular hypertrophy which did not increase up to 1 year of life. NP-deficient mice exposed to acute myocardial stress secondary to thoracic aortic constriction (TAC) had similar pathological myocardial remodeling but a significant increase in sudden death. We discovered that the NP-deficient mice are more susceptible to stress induced ventricular arrhythmias using both in vivo and ex vivo models. Mechanistically, deficiency of either ANP or BNP led to reduced myocardial cGMP levels and reduced phosphorylation of the cAMP response element-binding protein (CREBS133) transcriptional regulator. Selective CREB inhibition sensitized wild type hearts to stress induced ventricular arrhythmias. ANP and BNP regulate cardiomyocyte CREBS133 phosphorylation through a cGMP-dependent protein kinase 1 (PKG1) and p38 mitogen activated protein kinase (p38 MAPK) signaling cascade. CONCLUSIONS Our data show that ANP and BNP act in a non-redundant fashion to maintain myocardial cGMP levels to regulate cardiomyocyte p38 MAPK and CREB activity. Cardiac natriuretic peptide deficiency leads to a reduction in CREB signaling which sensitizes the heart to stress induced ventricular arrhythmias. TRANSLATIONAL PERSPECTIVE Our study found that ANP or BNP deficiency leads to increased sudden death and ventricular arrhythmias after acute myocardial stress in murine models. We discovered that ANP and BNP act in a non-redundant fashion to maintain myocardial cGMP levels and uncovered a unique role for these peptides in regulating cardiomyocyte p38 MAPK and CREB signaling through a cGMP-PKG1 pathway. Importantly, this signaling pathway was conserved in human cardiomyocytes. This study provides mechanistic insight into how modulating natriuretic peptide levels in human heart failure patients reduces sudden death and mortality.
Collapse
Affiliation(s)
- Eric J Hall
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Soumojit Pal
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute and Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Michael S Glennon
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute and Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Puneeth Shridhar
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute and Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Sidney L Satterfield
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute and Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Beth Weber
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute and Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Qinkun Zhang
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham Medical Center, Birmingham, AL, USA
| | - Guy Salama
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute and Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Hind Lal
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham Medical Center, Birmingham, AL, USA
| | - Jason R Becker
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute and Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
13
|
Silva-García O, Cortés-Vieyra R, Mendoza-Ambrosio FN, Ramírez-Galicia G, Baizabal-Aguirre VM. GSK3α: An Important Paralog in Neurodegenerative Disorders and Cancer. Biomolecules 2020; 10:E1683. [PMID: 33339170 PMCID: PMC7765659 DOI: 10.3390/biom10121683] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 11/29/2020] [Accepted: 11/30/2020] [Indexed: 12/18/2022] Open
Abstract
The biological activity of the enzyme glycogen synthase kinase-3 (GSK3) is fulfilled by two paralogs named GSK3α and GSK3β, which possess both redundancy and specific functions. The upregulated activity of these proteins is linked to the development of disorders such as neurodegenerative disorders (ND) and cancer. Although various chemical inhibitors of these enzymes restore the brain functions in models of ND such as Alzheimer's disease (AD), and reduce the proliferation and survival of cancer cells, the particular contribution of each paralog to these effects remains unclear as these molecules downregulate the activity of both paralogs with a similar efficacy. Moreover, given that GSK3 paralogs phosphorylate more than 100 substrates, the simultaneous inhibition of both enzymes has detrimental effects during long-term inhibition. Although the GSK3β kinase function has usually been taken as the global GSK3 activity, in the last few years, a growing interest in the study of GSK3α has emerged because several studies have recognized it as the main GSK3 paralog involved in a variety of diseases. This review summarizes the current biological evidence on the role of GSK3α in AD and various types of cancer. We also provide a discussion on some strategies that may lead to the design of the paralog-specific inhibition of GSK3α.
Collapse
Affiliation(s)
- Octavio Silva-García
- Departamento de Química Teórica, Universidad del Papaloapan, Oaxaca 68301, Mexico; (F.N.M.-A.); (G.R.-G.)
| | - Ricarda Cortés-Vieyra
- Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Michoacán 58000, Mexico;
| | | | - Guillermo Ramírez-Galicia
- Departamento de Química Teórica, Universidad del Papaloapan, Oaxaca 68301, Mexico; (F.N.M.-A.); (G.R.-G.)
| | - Víctor M. Baizabal-Aguirre
- Centro Multidisciplinario de Estudios en Biotecnología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás de Hidalgo, Michoacán 58893, Mexico
| |
Collapse
|
14
|
Beffagna G, Sommariva E, Bellin M. Mechanotransduction and Adrenergic Stimulation in Arrhythmogenic Cardiomyopathy: An Overview of in vitro and in vivo Models. Front Physiol 2020; 11:568535. [PMID: 33281612 PMCID: PMC7689294 DOI: 10.3389/fphys.2020.568535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 10/19/2020] [Indexed: 01/09/2023] Open
Abstract
Arrhythmogenic Cardiomyopathy (AC) is a rare inherited heart disease, manifesting with progressive myocardium degeneration and dysfunction, and life-threatening arrhythmic events that lead to sudden cardiac death. Despite genetic determinants, most of AC patients admitted to hospital are athletes or very physically active people, implying the existence of other disease-causing factors. It is recognized that AC phenotypes are enhanced and triggered by strenuous physical activity, while excessive mechanical stretch and load, and repetitive adrenergic stimulation are mechanisms influencing disease penetrance. Different approaches have been undertaken to recapitulate and study both mechanotransduction and adrenergic signaling in AC, including the use of in vitro cellular and tissue models, and the development of in vivo models (particularly rodents but more recently also zebrafish). However, it remains challenging to reproduce mechanical load stimuli and physical activity in laboratory experimental settings. Thus, more work to drive the innovation of advanced AC models is needed to recapitulate these subtle physiological influences. Here, we review the state-of-the-art in this field both in clinical and laboratory-based modeling scenarios. Specific attention will be focused on highlighting gaps in the knowledge and how they may be resolved by utilizing novel research methodology.
Collapse
Affiliation(s)
- Giorgia Beffagna
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Padua, Italy.,Department of Biology, University of Padua, Padua, Italy
| | - Elena Sommariva
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Milena Bellin
- Department of Biology, University of Padua, Padua, Italy.,Veneto Institute of Molecular Medicine, Padua, Italy.,Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
15
|
Mohammadpour H, Sarow JL, MacDonald CR, Chen GL, Qiu J, Sharma UC, Cao X, Herr MM, Hahn TE, Blazar BR, Repasky EA, McCarthy PL. β2-Adrenergic receptor activation on donor cells ameliorates acute GvHD. JCI Insight 2020; 5:137788. [PMID: 32437333 DOI: 10.1172/jci.insight.137788] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 05/13/2020] [Indexed: 12/12/2022] Open
Abstract
Acute graft versus host disease (aGvHD) remains a major impediment to successful allogeneic hematopoietic cell transplantation (allo-HCT). To solve this problem, a greater knowledge of factors that regulate the differentiation of donor T cells toward cytotoxic cells or Tregs is necessary. We report that the β2-adrenergic receptor (β2-AR) is critical for regulating this differentiation and that its manipulation can control aGvHD without impairing the graft-versus-tumor (GvT) effect. Donor T cell β2-AR expression and signaling is associated with decreased aGvHD when compared with recipients of β2-AR-/- donor T cells. We determined that β2-AR activation skewed CD4+ T cell differentiation in vitro and in vivo toward Tregs rather than the T helper 1 (Th1) phenotype. Treatment of allo-HCT recipients with a selective β2-agonist (bambuterol) ameliorated aGvHD severity. This was associated with increased Tregs, decreased cytotoxic T cells, and increased donor BM-derived myeloid-derived suppressor cells (MDSCs) in allogeneic and humanized xenogeneic aGvHD models. β2-AR signaling resulted in increased Treg generation through glycogen synthase kinase-3 activation. Bambuterol preserved the GvT effect by inducing NKG2D+ effector cells and central memory T cells. These data reveal how β-AR signaling can be targeted to ameliorate GvHD severity while preserving GvT effect.
Collapse
Affiliation(s)
| | | | | | - George L Chen
- Medicine, Transplant and Cellular Therapy Program, and
| | - Jingxin Qiu
- Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Umesh C Sharma
- Department of Medicine, Jacobs School of Medicine & Biomedical Sciences, Buffalo, New York, USA
| | - Xuefang Cao
- Department of Microbiology and Immunology, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland, USA
| | - Megan M Herr
- Medicine, Transplant and Cellular Therapy Program, and
| | | | - Bruce R Blazar
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | | | | |
Collapse
|
16
|
Singh AP, Umbarkar P, Guo Y, Force T, Gupte M, Lal H. Inhibition of GSK-3 to induce cardiomyocyte proliferation: a recipe for in situ cardiac regeneration. Cardiovasc Res 2020; 115:20-30. [PMID: 30321309 DOI: 10.1093/cvr/cvy255] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 10/09/2018] [Indexed: 01/03/2023] Open
Abstract
With an estimated 38 million current patients, heart failure (HF) is a leading cause of morbidity and mortality worldwide. Although the aetiology differs, HF is largely a disease of cardiomyocyte (CM) death or dysfunction. Due to the famously limited amount of regenerative capacity of the myocardium, the only viable option for advanced HF patients is cardiac transplantation; however, donor's hearts are in very short supply. Thus, novel regenerative strategies are urgently needed to reconstitute the injured hearts. Emerging data from our lab and others have elucidated that CM-specific deletion of glycogen synthase kinase (GSK)-3 family of kinases induces CM proliferation, and the degree of proliferation is amplified in the setting of cardiac stress. If this proliferation is sufficiently robust, one could induce meaningful regeneration without the need for delivering exogenous cells to the injured myocardium (i.e. cardiac regeneration in situ). Herein, we will discuss the emerging role of the GSK-3s in CM proliferation and differentiation, including their potential implications in cardiac regeneration. The underlying molecular interactions and cross-talk among signalling pathways will be discussed. We will also review the specificity and limitations of the available small molecule inhibitors targeting GSK-3 and their potential applications to stimulate the endogenous cardiac regenerative responses to repair the injured heart.
Collapse
Affiliation(s)
- Anand Prakash Singh
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, Suite PRB#348A, Nashville, TN, USA
| | - Prachi Umbarkar
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, Suite PRB#348A, Nashville, TN, USA
| | - Yuanjun Guo
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, Suite PRB#348A, Nashville, TN, USA.,Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Thomas Force
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, Suite PRB#348A, Nashville, TN, USA
| | - Manisha Gupte
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, Suite PRB#348A, Nashville, TN, USA
| | - Hind Lal
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, Suite PRB#348A, Nashville, TN, USA
| |
Collapse
|
17
|
Ahmad F, Woodgett JR. Emerging roles of GSK-3α in pathophysiology: Emphasis on cardio-metabolic disorders. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1867:118616. [PMID: 31785335 DOI: 10.1016/j.bbamcr.2019.118616] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 11/18/2019] [Accepted: 11/23/2019] [Indexed: 02/06/2023]
Abstract
Glycogen synthase kinase-3 (GSK-3) is a widely expressed serine/threonine kinase regulates a variety of cellular processes including proliferation, differentiation and death. Mammals harbor two structurally similar isoforms GSK-3α and β that have overlapping as well as unique functions. Of the two, GSK-3β has been studied (and reviewed) in far greater detail with analysis of GSK-3α often as an afterthought. It is now evident that systemic, chronic inhibition of either GSK-3β or both GSK-3α/β is not clinically feasible and if achieved would likely lead to adverse clinical conditions. Emerging evidence suggests important and specific roles for GSK-3α in fatty acid accumulation, insulin resistance, amyloid-β-protein precursor metabolism, atherosclerosis, cardiomyopathy, fibrosis, aging, fertility, and in a variety of cancers. Selective targeting of GSK-3α may present a novel therapeutic opportunity to alleviate a number of pathological conditions. In this review, we assess the evidence for roles of GSK-3α in a variety of pathophysiological settings.
Collapse
Affiliation(s)
- Firdos Ahmad
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates; Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates.
| | - James R Woodgett
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Canada
| |
Collapse
|
18
|
Zhang Z, Zhang Q, Lal H, Nam YJ. Generation of Nppa-tagBFP reporter knock-in mouse line for studying cardiac chamber specification. Genesis 2019; 57:e23294. [PMID: 30920727 DOI: 10.1002/dvg.23294] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 03/14/2019] [Accepted: 03/15/2019] [Indexed: 11/10/2022]
Abstract
Nppa is a cardiac hormone which plays critical roles in regulating salt-water balance. Its expression is restricted to the atria of the healthy post-natal heart. During heart development, spatio-temporal expression of Nppa is dynamically changed within the heart and becomes restricted to the atria upon birth. In contrast to its atrial specific expression after birth, Nppa is re-expressed in the adult ventricles in response to cardiac hypertrophy. To study cardiac chamber specification during development and pathological cardiac remodeling during heart disease, we generated a novel Nppa reporter mouse line by knocking-in a tagBFP reporter cassette into 3'-UTR of the Nppa gene without disrupting the endogenous gene. Our results demonstrated dynamic tagBFP expression in the developing heart, recapitulating the spatiotemporal expression pattern of endogenous Nppa. We also found that Nppa-tagBFP is induced in the ventricle during pathological remodeling. Taken together, Nppa-tagBFP reporter knock-in mouse model described in this article will serve as a valuable tool to study cardiac chamber specification during development as well as pathological cardiac remodeling.
Collapse
Affiliation(s)
- Zhentao Zhang
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee.,Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, Tennessee
| | - Qinkun Zhang
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Pharmacology, Vanderbilt University, Nashville, Tennessee
| | - Hind Lal
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Pharmacology, Vanderbilt University, Nashville, Tennessee
| | - Young-Jae Nam
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee.,Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
19
|
Ahmad F, Singh AP, Tomar D, Rahmani M, Zhang Q, Woodgett JR, Tilley DG, Lal H, Force T. Cardiomyocyte-GSK-3α promotes mPTP opening and heart failure in mice with chronic pressure overload. J Mol Cell Cardiol 2019; 130:65-75. [PMID: 30928428 DOI: 10.1016/j.yjmcc.2019.03.020] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 02/09/2019] [Accepted: 03/25/2019] [Indexed: 01/19/2023]
Abstract
Chronic pressure-overload (PO)- induced cardiomyopathy is one of the leading causes of left ventricular (LV) remodeling and heart failure. The role of the α isoform of glycogen synthase kinase-3 (GSK-3α) in PO-induced cardiac remodeling is unclear and its downstream molecular targets are largely unknown. To investigate the potential roles of GSK-3α, cardiomyocyte-specific GSK-3α conditional knockout (cKO) and control mice underwent trans-aortic constriction (TAC) or sham surgeries. Cardiac function in the cKOs and littermate controls declined equally up to 2 weeks of TAC. At 4 week, cKO animals retained concentric LV remodeling and showed significantly less decline in contractile function both at systole and diastole, vs. controls which remained same until the end of the study (6 wk). Histological analysis confirmed preservation of LV chamber and protection against TAC-induced cellular hypertrophy in the cKO. Consistent with attenuated hypertrophy, significantly lower level of cardiomyocyte apoptosis was observed in the cKO. Mechanistically, GSK-3α was found to regulate mitochondrial permeability transition pore (mPTP) opening and GSK-3α-deficient mitochondria showed delayed mPTP opening in response to Ca2+ overload. Consistently, overexpression of GSK-3α in cardiomyocytes resulted in elevated Bax expression, increased apoptosis, as well as a reduction of maximum respiration capacity and cell viability. Taken together, we show for the first time that GSK-3α regulates mPTP opening under pathological conditions, likely through Bax overexpression. Genetic ablation of cardiomyocyte GSK-3α protects against chronic PO-induced cardiomyopathy and adverse LV remodeling, and preserves contractile function. Selective inhibition of GSK-3α using isoform-specific inhibitors could be a viable therapeutic strategy to limit PO-induced heart failure.
Collapse
Affiliation(s)
- Firdos Ahmad
- College of Medicine and Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates.
| | - Anand P Singh
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Dhanendra Tomar
- Center for Translational Medicine, School of Medicine, Temple University, Philadelphia, PA, USA
| | - Mohamed Rahmani
- College of Medicine and Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Qinkun Zhang
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - James R Woodgett
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Douglas G Tilley
- Center for Translational Medicine, School of Medicine, Temple University, Philadelphia, PA, USA
| | - Hind Lal
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Thomas Force
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
20
|
Abstract
Glycogen synthase kinase-3 (GSK-3) is a cytoplasmic serine/threonine protein kinase which is known to regulate a variety of cellular processes through a number of signaling pathways important for cell proliferation, stem cell renewal, apoptosis and development. Although GSK-3 exists in a variety of tissues, this kinase plays very important roles in the heart to control its development through the formation of heart and cardiomyocyte proliferation. GSK-3 is also recognized as one of the main molecules that control cardiac hypertrophy and fibrosis. Therefore, GSK-3 could be an attractive target for the development of new drugs to cure cardiac diseases. The present review summarizes the roles of GSK-3 in the signaling pathways and the heart, and discusses the possibility of new drug development targeting this kinase.
Collapse
|
21
|
Yu SMW, Jean-Charles PY, Abraham DM, Kaur S, Gareri C, Mao L, Rockman HA, Shenoy SK. The deubiquitinase ubiquitin-specific protease 20 is a positive modulator of myocardial β 1-adrenergic receptor expression and signaling. J Biol Chem 2018; 294:2500-2518. [PMID: 30538132 DOI: 10.1074/jbc.ra118.004926] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 11/23/2018] [Indexed: 12/27/2022] Open
Abstract
Reversible ubiquitination of G protein-coupled receptors regulates their trafficking and signaling; whether deubiquitinases regulate myocardial β1-adrenergic receptors (β1ARs) is unknown. We report that ubiquitin-specific protease 20 (USP20) deubiquitinates and attenuates lysosomal trafficking of the β1AR. β1AR-induced phosphorylation of USP20 Ser-333 by protein kinase A-α (PKAα) was required for optimal USP20-mediated regulation of β1AR lysosomal trafficking. Both phosphomimetic (S333D) and phosphorylation-impaired (S333A) USP20 possess intrinsic deubiquitinase activity equivalent to WT activity. However, unlike USP20 WT and S333D, the S333A mutant associated poorly with the β1AR and failed to deubiquitinate the β1AR. USP20-KO mice showed normal baseline systolic function but impaired β1AR-induced contractility and relaxation. Dobutamine stimulation did not increase cAMP in USP20-KO left ventricles (LVs), whereas NKH477-induced adenylyl cyclase activity was equivalent to WT. The USP20 homolog USP33, which shares redundant roles with USP20, had no effect on β1AR ubiquitination, but USP33 was up-regulated in USP20-KO hearts suggesting compensatory regulation. Myocardial β1AR expression in USP20-KO was drastically reduced, whereas β2AR expression was maintained as determined by radioligand binding in LV sarcolemmal membranes. Phospho-USP20 was significantly increased in LVs of wildtype (WT) mice after a 1-week catecholamine infusion and a 2-week chronic pressure overload induced by transverse aortic constriction (TAC). Phospho-USP20 was undetectable in β1AR KO mice subjected to TAC, suggesting a role for USP20 phosphorylation in cardiac response to pressure overload. We conclude that USP20 regulates β1AR signaling in vitro and in vivo Additionally, β1AR-induced USP20 phosphorylation may serve as a feed-forward mechanism to stabilize β1AR expression and signaling during pathological insults to the myocardium.
Collapse
Affiliation(s)
- Samuel Mon-Wei Yu
- From the Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, North Carolina 27710
| | - Pierre-Yves Jean-Charles
- From the Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, North Carolina 27710
| | - Dennis M Abraham
- From the Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, North Carolina 27710
| | - Suneet Kaur
- From the Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, North Carolina 27710
| | - Clarice Gareri
- From the Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, North Carolina 27710
| | - Lan Mao
- From the Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, North Carolina 27710
| | - Howard A Rockman
- From the Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, North Carolina 27710
| | - Sudha K Shenoy
- From the Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, North Carolina 27710
| |
Collapse
|
22
|
Zhou J, Ahmad F, Lal H, Force T. Response by Zhou et al to Letter Regarding Article, "Loss of Adult Cardiac Myocyte GSK-3 Leads to Mitotic Catastrophe Resulting in Fatal Dilated Cardiomyopathy". Circ Res 2018; 119:e29-e30. [PMID: 27390340 DOI: 10.1161/circresaha.116.309093] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Jibin Zhou
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA
| | - Firdos Ahmad
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Hind Lal
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Thomas Force
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
23
|
Dey S, Goswami S, Eisa A, Bhattacharjee R, Brothag C, Kline D, Vijayaraghavan S. Cyclic AMP and glycogen synthase kinase 3 form a regulatory loop in spermatozoa. J Cell Physiol 2018; 233:7239-7252. [PMID: 29574946 DOI: 10.1002/jcp.26557] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 02/20/2018] [Indexed: 12/17/2022]
Abstract
The multifaceted glycogen synthase kinase (GSK3) has an essential role in sperm and male fertility. Since cyclic AMP (cAMP) plays an important role in sperm function, we investigated whether GSK3 and cAMP pathways may be interrelated. We used GSK3 and soluble adenylyl cyclase (sAC) knockout mice and pharmacological modulators to examine this relationship. Intracellular cAMP levels were found to be significantly lower in sperm lacking GSK3α or GSK3β. A similar outcome was observed when sperm cells were treated with SB216763, a GSK3 inhibitor. This reduction of cAMP levels was not due to an effect on sperm adenylyl cyclase but was caused by elevated phosphodiesterase (PDE) activity. The PDE4 inhibitor RS25344 or the general PDE inhibitor IBMX could restore cAMP levels in sperm lacking GSK3α or β-isoform. PDE activity assay also showed that hyperactivated PDE4 contributes in lowering of cAMP levels in GSK3α null sperm suggesting that in wild-type sperm PDE4 activity is kept in check by GSK3. Conversely, PKA being triggered by cAMP, affected GSK3 activity through increasing its phosphorylation. Increased GSK3 phosphorylation also occurred by inhibition of sperm specific protein phosphatase type 1, PP1γ2. The relationship between cAMP, GSK3, and PP1γ2 activities was also confirmed in sperm from sAC null mice. Pull-down assay using recombinant PP1γ2 indicated that PKA, GSK3, and PP1γ2 could exist as a complex. Pharmacological inhibition of GSK3 in mature spermatozoa resulted in significantly reduced fertilization of eggs in vitro. Our results show that cAMP, PKA, and GSK3 are interrelated in regulation of sperm function.
Collapse
Affiliation(s)
- Souvik Dey
- Department of Biological Sciences, Kent State University, Kent, Ohio
| | - Suranjana Goswami
- Department of Biological Sciences, Kent State University, Kent, Ohio
| | - Alaa Eisa
- School of Biomedical Sciences, Kent State University, Kent, Ohio
| | | | - Cameron Brothag
- Department of Biological Sciences, Kent State University, Kent, Ohio
| | - Douglas Kline
- Department of Biological Sciences, Kent State University, Kent, Ohio
| | | |
Collapse
|
24
|
Cardiomyocyte-specific deletion of GSK-3β leads to cardiac dysfunction in a diet induced obesity model. Int J Cardiol 2018; 259:145-152. [PMID: 29398139 DOI: 10.1016/j.ijcard.2018.01.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 12/19/2017] [Accepted: 01/02/2018] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND RATIONALE Obesity, an independent risk factor for the development of myocardial diseases is a growing healthcare problem worldwide. It's well established that GSK-3β is critical to cardiac pathophysiology. However, the role cardiomyocyte (CM) GSK-3β in diet-induced cardiac dysfunction is unknown. METHODS CM-specific GSK-3β knockout (CM-GSK-3β-KO) and littermate controls (WT) mice were fed either a control diet (CD) or high-fat diet (HFD) for 55weeks. Cardiac function was assessed by transthoracic echocardiography. RESULTS At baseline, body weights and cardiac function were comparable between the WT and CM-GSK-3β-KOs. However, HFD-fed CM-GSK-3β-KO mice developed severe cardiac dysfunction. Consistently, both heart weight/tibia length and lung weight/tibia length were significantly elevated in the HFD-fed CM-GSK-3β-KO mice. The impaired cardiac function and adverse ventricular remodeling in the CM-GSK-3β-KOs were independent of body weight or the lean/fat mass composition as HFD-fed CM-GSK-3β-KO and controls demonstrated comparable body weight and body masses. At the molecular level, on a CD, CM-GSK-3α compensated for the loss of CM-GSK-3β, as evident by significantly reduced GSK-3αs21 phosphorylation (activation) resulting in a preserved canonical β-catenin ubiquitination pathway and cardiac function. However, this protective compensatory mechanism is lost with HFD, leading to excessive accumulation of β-catenin in HFD-fed CM-GSK-3β-KO hearts, resulting in adverse ventricular remodeling and cardiac dysfunction. CONCLUSION In summary, these results suggest that cardiac GSK-3β is crucial to protect against obesity-induced adverse ventricular remodeling and cardiac dysfunction.
Collapse
|
25
|
Chen X, Wang R, Liu X, Wu Y, Zhou T, Yang Y, Perez A, Chen YC, Hu L, Chadarevian JP, Assadieskandar A, Zhang C, Ying QL. A Chemical-Genetic Approach Reveals the Distinct Roles of GSK3α and GSK3β in Regulating Embryonic Stem Cell Fate. Dev Cell 2018; 43:563-576.e4. [PMID: 29207259 DOI: 10.1016/j.devcel.2017.11.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 08/30/2017] [Accepted: 11/06/2017] [Indexed: 11/30/2022]
Abstract
Glycogen synthase kinase 3 (GSK3) plays a central role in diverse cellular processes. GSK3 has two mammalian isozymes, GSK3α and GSK3β, whose functions remain ill-defined because of a lack of inhibitors that can distinguish between the two highly homologous isozymes. Here, we show that GSK3α and GSK3β can be selectively inhibited in mouse embryonic stem cells (ESCs) using a chemical-genetic approach. Selective inhibition of GSK3β is sufficient to maintain mouse ESC self-renewal, whereas GSK3α inhibition promotes mouse ESC differentiation toward neural lineages. Genome-wide transcriptional analysis reveals that GSK3α and GSK3β have distinct sets of downstream targets. Furthermore, selective inhibition of individual GSK3 isozymes yields distinct phenotypes from gene deletion, highlighting the power of the chemical-genetic approach in dissecting kinase catalytic functions from the protein's scaffolding functions. Our study opens new avenues for defining GSK3 isozyme-specific functions in various cellular processes.
Collapse
Affiliation(s)
- Xi Chen
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Ruizhe Wang
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Xu Liu
- Loker Hydrocarbon Research Institute & Department of Chemistry, University of Southern California, Los Angeles, CA 90089, USA
| | - Yongming Wu
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Tao Zhou
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Yujia Yang
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Andrew Perez
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Ying-Chu Chen
- Loker Hydrocarbon Research Institute & Department of Chemistry, University of Southern California, Los Angeles, CA 90089, USA
| | - Liang Hu
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Jean Paul Chadarevian
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Amir Assadieskandar
- Loker Hydrocarbon Research Institute & Department of Chemistry, University of Southern California, Los Angeles, CA 90089, USA
| | - Chao Zhang
- Loker Hydrocarbon Research Institute & Department of Chemistry, University of Southern California, Los Angeles, CA 90089, USA.
| | - Qi-Long Ying
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
26
|
GSK-3 Inhibitors: Anti-Diabetic Treatment Associated with Cardiac Risk? : Editorial to: "The Impact of Chronic Glycogen Synthase Kinase-3 Inhibition on Remodeling of Normal and Pre-Diabetic Rat Hearts." by Barbara Huisamen et al. Cardiovasc Drugs Ther 2017; 30:233-5. [PMID: 27311575 DOI: 10.1007/s10557-016-6669-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
27
|
Fujita A, Takahashi-Yanaga F, Morimoto S, Yoshihara T, Arioka M, Igawa K, Tomooka K, Hoka S, Sasaguri T. 2,5-Dimethylcelecoxib prevents pressure-induced left ventricular remodeling through GSK-3 activation. Hypertens Res 2016; 40:130-139. [PMID: 27628899 DOI: 10.1038/hr.2016.122] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 06/15/2016] [Accepted: 08/01/2016] [Indexed: 01/01/2023]
Abstract
Glycogen synthase kinase-3 (GSK-3) is a crucial regulator of cardiac hypertrophy. We previously reported that 2,5-dimethylcelecoxib (DM-celecoxib), a celecoxib derivative unable to inhibit cyclooxygenase-2, prevented cardiac remodeling by activating GSK-3, resulting in lifespan prolongation in a mouse model of genetic dilated cardiomyopathy. In the present study, we investigated whether DM-celecoxib can also prevent pressure-induced cardiac remodeling and heart failure, elicited by transverse aortic constriction (TAC). Before testing the effects of DM-celecoxib, we compared the effects of TAC on the hearts of wild-type and GSK-3β hetero-deficient (GSK-3β+/-) mice to determine the role of GSK-3 in cardiac remodeling and heart failure. GSK-3β+/- mouse hearts exhibited more severe hypertrophy, which was characterized by accelerated interstitial fibrosis, than wild-type mouse hearts after TAC, suggesting that reduced GSK-3β activity aggravates pressure-induced left ventricular remodeling. We subsequently examined the effects of DM-celecoxib on TAC-induced cardiac remodeling. DM-celecoxib inhibited left ventricular systolic functional deterioration, and prevented left ventricular hypertrophy and fibrosis. It also activated GSK-3α and β by inhibiting Akt, suppressing the activity of β-catenin and nuclear factor of activated T-cells and thereby decreasing the expression of the Wnt/β-catenin target gene products fibronectin and matrix metalloproteinase-2. These results suggest that DM-celecoxib is clinically useful for treating pressure-induced heart diseases.
Collapse
Affiliation(s)
- Ai Fujita
- Department of Clinical Pharmacology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Anesthesia and Critical Care Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Fumi Takahashi-Yanaga
- Department of Clinical Pharmacology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.,Global Medical Science Education Unit, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Sachio Morimoto
- Department of Clinical Pharmacology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tatsuya Yoshihara
- Department of Clinical Pharmacology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masaki Arioka
- Department of Clinical Pharmacology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazunobu Igawa
- Department of Molecular and Material Science, Institute for Materials Chemistry and Engineering, Kyushu University, Fukuoka, Japan
| | - Katsuhiko Tomooka
- Department of Molecular and Material Science, Institute for Materials Chemistry and Engineering, Kyushu University, Fukuoka, Japan
| | - Sumio Hoka
- Department of Anesthesia and Critical Care Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Toshiyuki Sasaguri
- Department of Clinical Pharmacology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
28
|
Kakade VR, Tao S, Rajagopal M, Zhou X, Li X, Yu ASL, Calvet JP, Pandey P, Rao R. A cAMP and CREB-mediated feed-forward mechanism regulates GSK3β in polycystic kidney disease. J Mol Cell Biol 2016; 8:464-476. [PMID: 27190311 DOI: 10.1093/jmcb/mjw022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 02/05/2016] [Accepted: 02/28/2016] [Indexed: 12/12/2022] Open
Abstract
Glycogen synthase kinase 3β (GSK3β), a serine/threonine protein kinase, is commonly known to be regulated at the level of its activity. However, in some diseases including polycystic kidney disease (PKD), GSK3β expression is increased and plays a pathophysiological role. The current studies aimed to determine the mechanism for the increased GSK3β expression in PKD and its significance to disease progression. In mouse models of PKD, increases in renal GSK3β corresponded with increases in renal cAMP levels and disease progression. In vivo and in vitro studies revealed that GSK3β is a cAMP-responsive gene, and elevated cAMP levels, as seen in PKD, can increase GSK3β expression. In normal mice, vasopressin signaling induced by water deprivation increased GSK3β expression, which decreased following rehydration. Examination of the GSK3β promoter revealed five potential binding sites for the transcription factor, cAMP response element binding protein (CREB). CREB was found to bind to GSK3β promoter and essential for cAMP-mediated regulation of GSK3β. Importantly, this regulation was demonstrated to be part of a feed-forward loop in which cAMP through CREB regulates GSK3β expression, and GSK3β in turn positively regulates cAMP generation. GSK3β or CREB inhibition reduced transepithelial fluid secretion and cyst expansion in vitro Thus, disruption at any point of this destructive cycle may be therapeutically useful to reduce cyst expansion and preserve renal function in PKD.
Collapse
Affiliation(s)
- Vijayakumar R Kakade
- The Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Shixin Tao
- The Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Madhumitha Rajagopal
- The Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Xia Zhou
- The Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Xiaogang Li
- The Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Alan S L Yu
- The Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - James P Calvet
- The Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Pankaj Pandey
- The Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Reena Rao
- The Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
29
|
Zhou J, Ahmad F, Parikh S, Hoffman NE, Rajan S, Verma VK, Song J, Yuan A, Shanmughapriya S, Guo Y, Gao E, Koch W, Woodgett JR, Madesh M, Kishore R, Lal H, Force T. Loss of Adult Cardiac Myocyte GSK-3 Leads to Mitotic Catastrophe Resulting in Fatal Dilated Cardiomyopathy. Circ Res 2016; 118:1208-22. [PMID: 26976650 DOI: 10.1161/circresaha.116.308544] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 03/14/2016] [Indexed: 11/16/2022]
Abstract
RATIONALE Cardiac myocyte-specific deletion of either glycogen synthase kinase (GSK)-3α and GSK-3β leads to cardiac protection after myocardial infarction, suggesting that deletion of both isoforms may provide synergistic protection. This is an important consideration because of the fact that all GSK-3-targeted drugs, including the drugs already in clinical trial target both isoforms of GSK-3, and none are isoform specific. OBJECTIVE To identify the consequences of combined deletion of cardiac myocyte GSK-3α and GSK-3β in heart function. METHODS AND RESULTS We generated tamoxifen-inducible cardiac myocyte-specific mice lacking both GSK-3 isoforms (double knockout). We unexpectedly found that cardiac myocyte GSK-3 is essential for cardiac homeostasis and overall survival. Serial echocardiographic analysis reveals that within 2 weeks of tamoxifen treatment, double-knockout hearts leads to excessive dilatative remodeling and ventricular dysfunction. Further experimentation with isolated adult cardiac myocytes and fibroblasts from double-knockout implicated cardiac myocytes intrinsic factors responsible for observed phenotype. Mechanistically, loss of GSK-3 in adult cardiac myocytes resulted in induction of mitotic catastrophe, a previously unreported event in cardiac myocytes. Double-knockout cardiac myocytes showed cell cycle progression resulting in increased DNA content and multinucleation. However, increased cell cycle activity was rivaled by marked activation of DNA damage, cell cycle checkpoint activation, and mitotic catastrophe-induced apoptotic cell death. Importantly, mitotic catastrophe was also confirmed in isolated adult cardiac myocytes. CONCLUSIONS Together, our findings suggest that cardiac myocyte GSK-3 is required to maintain normal cardiac homeostasis, and its loss is incompatible with life because of cell cycle dysregulation that ultimately results in a severe fatal dilated cardiomyopathy.
Collapse
Affiliation(s)
- Jibin Zhou
- From the Division of Cardiovascular Medicine (F.A., V.K.V., Y.G., H.L., T.F.) and Department of Pharmacology (S.P., Y.G.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (J.Z., N.E.H., S.R., J.S., A.Y., S.S., E.G., W.K., M.M., R.K.); and Department of Medical Biophysics, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada (J.R.W.)
| | - Firdos Ahmad
- From the Division of Cardiovascular Medicine (F.A., V.K.V., Y.G., H.L., T.F.) and Department of Pharmacology (S.P., Y.G.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (J.Z., N.E.H., S.R., J.S., A.Y., S.S., E.G., W.K., M.M., R.K.); and Department of Medical Biophysics, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada (J.R.W.)
| | - Shan Parikh
- From the Division of Cardiovascular Medicine (F.A., V.K.V., Y.G., H.L., T.F.) and Department of Pharmacology (S.P., Y.G.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (J.Z., N.E.H., S.R., J.S., A.Y., S.S., E.G., W.K., M.M., R.K.); and Department of Medical Biophysics, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada (J.R.W.)
| | - Nichole E Hoffman
- From the Division of Cardiovascular Medicine (F.A., V.K.V., Y.G., H.L., T.F.) and Department of Pharmacology (S.P., Y.G.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (J.Z., N.E.H., S.R., J.S., A.Y., S.S., E.G., W.K., M.M., R.K.); and Department of Medical Biophysics, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada (J.R.W.)
| | - Sudarsan Rajan
- From the Division of Cardiovascular Medicine (F.A., V.K.V., Y.G., H.L., T.F.) and Department of Pharmacology (S.P., Y.G.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (J.Z., N.E.H., S.R., J.S., A.Y., S.S., E.G., W.K., M.M., R.K.); and Department of Medical Biophysics, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada (J.R.W.)
| | - Vipin K Verma
- From the Division of Cardiovascular Medicine (F.A., V.K.V., Y.G., H.L., T.F.) and Department of Pharmacology (S.P., Y.G.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (J.Z., N.E.H., S.R., J.S., A.Y., S.S., E.G., W.K., M.M., R.K.); and Department of Medical Biophysics, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada (J.R.W.)
| | - Jianliang Song
- From the Division of Cardiovascular Medicine (F.A., V.K.V., Y.G., H.L., T.F.) and Department of Pharmacology (S.P., Y.G.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (J.Z., N.E.H., S.R., J.S., A.Y., S.S., E.G., W.K., M.M., R.K.); and Department of Medical Biophysics, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada (J.R.W.)
| | - Ancai Yuan
- From the Division of Cardiovascular Medicine (F.A., V.K.V., Y.G., H.L., T.F.) and Department of Pharmacology (S.P., Y.G.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (J.Z., N.E.H., S.R., J.S., A.Y., S.S., E.G., W.K., M.M., R.K.); and Department of Medical Biophysics, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada (J.R.W.)
| | - Santhanam Shanmughapriya
- From the Division of Cardiovascular Medicine (F.A., V.K.V., Y.G., H.L., T.F.) and Department of Pharmacology (S.P., Y.G.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (J.Z., N.E.H., S.R., J.S., A.Y., S.S., E.G., W.K., M.M., R.K.); and Department of Medical Biophysics, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada (J.R.W.)
| | - Yuanjun Guo
- From the Division of Cardiovascular Medicine (F.A., V.K.V., Y.G., H.L., T.F.) and Department of Pharmacology (S.P., Y.G.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (J.Z., N.E.H., S.R., J.S., A.Y., S.S., E.G., W.K., M.M., R.K.); and Department of Medical Biophysics, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada (J.R.W.)
| | - Erhe Gao
- From the Division of Cardiovascular Medicine (F.A., V.K.V., Y.G., H.L., T.F.) and Department of Pharmacology (S.P., Y.G.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (J.Z., N.E.H., S.R., J.S., A.Y., S.S., E.G., W.K., M.M., R.K.); and Department of Medical Biophysics, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada (J.R.W.)
| | - Walter Koch
- From the Division of Cardiovascular Medicine (F.A., V.K.V., Y.G., H.L., T.F.) and Department of Pharmacology (S.P., Y.G.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (J.Z., N.E.H., S.R., J.S., A.Y., S.S., E.G., W.K., M.M., R.K.); and Department of Medical Biophysics, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada (J.R.W.)
| | - James R Woodgett
- From the Division of Cardiovascular Medicine (F.A., V.K.V., Y.G., H.L., T.F.) and Department of Pharmacology (S.P., Y.G.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (J.Z., N.E.H., S.R., J.S., A.Y., S.S., E.G., W.K., M.M., R.K.); and Department of Medical Biophysics, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada (J.R.W.)
| | - Muniswamy Madesh
- From the Division of Cardiovascular Medicine (F.A., V.K.V., Y.G., H.L., T.F.) and Department of Pharmacology (S.P., Y.G.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (J.Z., N.E.H., S.R., J.S., A.Y., S.S., E.G., W.K., M.M., R.K.); and Department of Medical Biophysics, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada (J.R.W.)
| | - Raj Kishore
- From the Division of Cardiovascular Medicine (F.A., V.K.V., Y.G., H.L., T.F.) and Department of Pharmacology (S.P., Y.G.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (J.Z., N.E.H., S.R., J.S., A.Y., S.S., E.G., W.K., M.M., R.K.); and Department of Medical Biophysics, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada (J.R.W.)
| | - Hind Lal
- From the Division of Cardiovascular Medicine (F.A., V.K.V., Y.G., H.L., T.F.) and Department of Pharmacology (S.P., Y.G.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (J.Z., N.E.H., S.R., J.S., A.Y., S.S., E.G., W.K., M.M., R.K.); and Department of Medical Biophysics, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada (J.R.W.).
| | - Thomas Force
- From the Division of Cardiovascular Medicine (F.A., V.K.V., Y.G., H.L., T.F.) and Department of Pharmacology (S.P., Y.G.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (J.Z., N.E.H., S.R., J.S., A.Y., S.S., E.G., W.K., M.M., R.K.); and Department of Medical Biophysics, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada (J.R.W.).
| |
Collapse
|
30
|
Sallam K, Li Y, Sager PT, Houser SR, Wu JC. Finding the rhythm of sudden cardiac death: new opportunities using induced pluripotent stem cell-derived cardiomyocytes. Circ Res 2015; 116:1989-2004. [PMID: 26044252 DOI: 10.1161/circresaha.116.304494] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Sudden cardiac death is a common cause of death in patients with structural heart disease, genetic mutations, or acquired disorders affecting cardiac ion channels. A wide range of platforms exist to model and study disorders associated with sudden cardiac death. Human clinical studies are cumbersome and are thwarted by the extent of investigation that can be performed on human subjects. Animal models are limited by their degree of homology to human cardiac electrophysiology, including ion channel expression. Most commonly used cellular models are cellular transfection models, which are able to mimic the expression of a single-ion channel offering incomplete insight into changes of the action potential profile. Induced pluripotent stem cell-derived cardiomyocytes resemble, but are not identical, adult human cardiomyocytes and provide a new platform for studying arrhythmic disorders leading to sudden cardiac death. A variety of platforms exist to phenotype cellular models, including conventional and automated patch clamp, multielectrode array, and computational modeling. Induced pluripotent stem cell-derived cardiomyocytes have been used to study long QT syndrome, catecholaminergic polymorphic ventricular tachycardia, hypertrophic cardiomyopathy, and other hereditary cardiac disorders. Although induced pluripotent stem cell-derived cardiomyocytes are distinct from adult cardiomyocytes, they provide a robust platform to advance the science and clinical care of sudden cardiac death.
Collapse
Affiliation(s)
- Karim Sallam
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (K.S., Y.L., P.T.S., J.C.W.), Institute of Stem Cell Biology and Regenerative Medicine (K.S., Y.L., J.C.W.), Stanford University School of Medicine, CA; and Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (S.R.H.)
| | - Yingxin Li
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (K.S., Y.L., P.T.S., J.C.W.), Institute of Stem Cell Biology and Regenerative Medicine (K.S., Y.L., J.C.W.), Stanford University School of Medicine, CA; and Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (S.R.H.)
| | - Philip T Sager
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (K.S., Y.L., P.T.S., J.C.W.), Institute of Stem Cell Biology and Regenerative Medicine (K.S., Y.L., J.C.W.), Stanford University School of Medicine, CA; and Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (S.R.H.)
| | - Steven R Houser
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (K.S., Y.L., P.T.S., J.C.W.), Institute of Stem Cell Biology and Regenerative Medicine (K.S., Y.L., J.C.W.), Stanford University School of Medicine, CA; and Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (S.R.H.).
| | - Joseph C Wu
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (K.S., Y.L., P.T.S., J.C.W.), Institute of Stem Cell Biology and Regenerative Medicine (K.S., Y.L., J.C.W.), Stanford University School of Medicine, CA; and Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (S.R.H.).
| |
Collapse
|
31
|
Abstract
It is well established that cardiac remodeling plays a pivotal role in the development of heart failure, a leading cause of death worldwide. Meanwhile, sympathetic hyperactivity is an important factor in inducing cardiac remodeling. Therefore, an in-depth understanding of beta-adrenoceptor signaling pathways would help to find better ways to reverse the adverse remodeling. Here, we reviewed five pathways, namely mitogen-activated protein kinase signaling, Gs-AC-cAMP signaling, Ca(2+)-calcineurin-NFAT/CaMKII-HDACs signaling, PI3K signaling and beta-3 adrenergic signaling, in cardiac remodeling. Furthermore, we constructed a cardiac-remodeling-specific regulatory network including miRNA, transcription factors and target genes within the five pathways. Both experimental and clinical studies have documented beneficial effects of beta blockers in cardiac remodeling; nevertheless, different blockers show different extent of therapeutic effect. Exploration of the underlying mechanisms could help developing more effective drugs. Current evidence of treatment effect of beta blockers in remodeling was also reviewed based upon information from experimental data and clinical trials. We further discussed the mechanism of how beta blockers work and why some beta blockers are more potent than others in treating cardiac remodeling within the framework of cardiac remodeling network.
Collapse
|
32
|
Kobayashi S, Nakamura TY, Wakabayashi S. Calcineurin B homologous protein 3 negatively regulates cardiomyocyte hypertrophy via inhibition of glycogen synthase kinase 3 phosphorylation. J Mol Cell Cardiol 2015; 84:133-42. [PMID: 25935310 DOI: 10.1016/j.yjmcc.2015.04.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 04/24/2015] [Accepted: 04/25/2015] [Indexed: 10/23/2022]
Abstract
Cardiac hypertrophy is a leading cause of serious heart diseases. Although many signaling molecules are involved in hypertrophy, the functions of some proteins in this process are still unknown. Calcineurin B homologous protein 3 (CHP3)/tescalcin is an EF-hand Ca(2+)-binding protein that is abundantly expressed in the heart; however, the function of CHP3 is unclear. Here, we aimed to identify the cardiac functions of CHP3. CHP3 was expressed in hearts at a wide range of developmental stages and was specifically detected in neonatal rat ventricular myocytes (NRVMs) but not in cardiac fibroblasts in culture. Moreover, knockdown of CHP3 expression using adenoviral-based RNA interference in NRVMs resulted in enlargement of cardiomyocyte size, concomitant with increased expression of a pathological hypertrophy marker ANP. This same treatment elevated glycogen synthase kinase (GSK3α/β) phosphorylation, which is known to inhibit GSK3 function. In contrast, CHP3 overexpression blocked the insulin-induced phosphorylation of GSK3α/β without affecting the phosphorylation of Akt, which is an upstream kinase of GSK3α/β, in HEK293 cells, and it inhibited both IGF-1-induced phosphorylation of GSK3β and cardiomyocyte hypertrophy in NRVMs. Co-immunoprecipitation experiments revealed that GSK3β interacted with CHP3. However, a Ca(2+)-binding-defective mutation of CHP3 (CHP3-D123A) also interacted with GSK3β and had the same inhibitory effect on GSK3α/β phosphorylation, suggesting that the action of CHP3 was independent of Ca(2+). These findings suggest that CHP3 functions as a novel negative regulator of cardiomyocyte hypertrophy via inhibition of GSK3α/β phosphorylation and subsequent enzymatic activation of GSK3α/β.
Collapse
Affiliation(s)
- Soushi Kobayashi
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Fujishirodai 5-7-1, Suita, Osaka 565-8565, Japan
| | - Tomoe Y Nakamura
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Fujishirodai 5-7-1, Suita, Osaka 565-8565, Japan
| | - Shigeo Wakabayashi
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Fujishirodai 5-7-1, Suita, Osaka 565-8565, Japan.
| |
Collapse
|
33
|
Monteiro da Rocha A, Ding J, Slawny N, Wolf AM, Smith GD. Loss of glycogen synthase kinase 3 isoforms during murine oocyte growth induces offspring cardiac dysfunction. Biol Reprod 2015; 92:127. [PMID: 25833158 DOI: 10.1095/biolreprod.115.128181] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 03/19/2015] [Indexed: 12/22/2022] Open
Abstract
Glycogen synthase kinase-3 (GSK3) is a constitutively active serine threonine kinase with 1) two isoforms (GSK3A and GSK3B) that have unique and overlapping functions, 2) multiple molecular intracellular mechanisms that involve phosphorylation of diverse substrates, and 3) implications in pathogenesis of many diseases. Insulin causes phosphorylation and inactivation of GSK3 and mammalian oocytes have a functional insulin-signaling pathway whereby prolonged elevated insulin during follicle/oocyte development causes GSK3 hyperphosphorylation, reduced GSK3 activity, and altered oocyte chromatin remodeling. Periconceptional diabetes and chronic hyperinsulinemia are associated with congenital malformations and onset of adult diseases of cardiovascular origin. Objectives were to produce transgenic mice with individual or concomitant loss of GSK3A and/or GSK3B and investigate the in vivo role of oocyte GSK3 on fertility, fetal development, and offspring health. Wild-type males bred to females with individual or concomitant loss of oocyte GSK3 isoforms did not have reduced fertility. However, concomitant loss of GSK3A and GSK3B in the oocyte significantly increased neonatal death rate due to congestive heart failure secondary to ventricular hyperplasia. Individual loss of oocyte GSK3A or GSK3B did not induce this lethal phenotype. In conclusion, absence of oocyte GSK3 in the periconceptional period does not alter fertility yet causes offspring cardiac hyperplasia, cardiovascular defects, and significant neonatal death. These results support a developmental mechanism by which periconceptional hyperinsulinemia associated with maternal metabolic syndrome, obesity, and/or diabetes can act on the oocyte and affect offspring cardiovascular development, function, and congenital heart malformation.
Collapse
Affiliation(s)
| | - Jun Ding
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan
| | - Nicole Slawny
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan
| | - Amber M Wolf
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Gary D Smith
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan Department of Urology, University of Michigan, Ann Arbor, Michigan Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
34
|
Abstract
Glycogen synthase kinase-3 (GSK-3) is one of the few signaling molecules that regulate a truly astonishing number of critical intracellular signaling pathways. It has been implicated in several diseases including heart failure, bipolar disorder, diabetes mellitus, Alzheimer disease, aging, inflammation, and cancer. Furthermore, a recent clinical trial has validated the feasibility of targeting GSK-3 with small molecule inhibitors for human diseases. In the current review, we will focus on its expanding role in the heart, concentrating primarily on recent studies that have used cardiomyocyte- and fibroblast-specific conditional gene deletion in mouse models. We will highlight the role of the GSK-3 isoforms in various pathological conditions including myocardial aging, ischemic injury, myocardial fibrosis, and cardiomyocyte proliferation. We will discuss our recent findings that deletion of GSK-3α specifically in cardiomyocytes attenuates ventricular remodeling and cardiac dysfunction after myocardial infarction by limiting scar expansion and promoting cardiomyocyte proliferation. The recent emergence of GSK-3β as a regulator of myocardial fibrosis will also be discussed. We will review our recent findings that specific deletion of GSK-3β in cardiac fibroblasts leads to fibrogenesis, left ventricular dysfunction, and excessive scarring in the ischemic heart. Finally, we will examine the underlying mechanisms that drive the aberrant myocardial fibrosis in the models in which GSK-3β is specifically deleted in cardiac fibroblasts. We will summarize these recent results and offer explanations, whenever possible, and hypotheses when not. For these studies we will rely heavily on our models and those of others to reconcile some of the apparent inconsistencies in the literature.
Collapse
Affiliation(s)
- Hind Lal
- From the Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN (H.L., F.A., T.F.); and Department of Medical Biophysics, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada (J.W.).
| | - Firdos Ahmad
- From the Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN (H.L., F.A., T.F.); and Department of Medical Biophysics, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada (J.W.)
| | - James Woodgett
- From the Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN (H.L., F.A., T.F.); and Department of Medical Biophysics, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada (J.W.)
| | - Thomas Force
- From the Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN (H.L., F.A., T.F.); and Department of Medical Biophysics, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada (J.W.).
| |
Collapse
|
35
|
Nørregaard R, Tao S, Nilsson L, Woodgett JR, Kakade V, Yu ASL, Howard C, Rao R. Glycogen synthase kinase 3α regulates urine concentrating mechanism in mice. Am J Physiol Renal Physiol 2015; 308:F650-60. [PMID: 25608967 DOI: 10.1152/ajprenal.00516.2014] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In mammals, glycogen synthase kinase (GSK)3 comprises GSK3α and GSK3β isoforms. GSK3β has been shown to play a role in the ability of kidneys to concentrate urine by regulating vasopressin-mediated water permeability of collecting ducts, whereas the role of GSK3α has yet to be discerned. To investigate the role of GSK3α in urine concentration, we compared GSK3α knockout (GSK3αKO) mice with wild-type (WT) littermates. Under normal conditions, GSK3αKO mice had higher water intake and urine output. GSK3αKO mice also showed reduced urine osmolality and aquaporin-2 levels but higher urinary vasopressin. When water deprived, they failed to concentrate their urine to the same level as WT littermates. The addition of 1-desamino-8-d-arginine vasopressin to isolated inner medullary collecting ducts increased the cAMP response in WT mice, but this response was reduced in GSK3αKO mice, suggesting reduced responsiveness to vasopressin. Gene silencing of GSK3α in mpkCCD cells also reduced forskolin-induced aquaporin-2 expression. When treated with LiCl, an isoform nonselective inhibitor of GSK3 and known inducer of polyuria, WT mice developed significant polyuria within 6 days. However, in GSK3αKO mice, the polyuric response was markedly reduced. This study demonstrates, for the first time, that GSK3α could play a crucial role in renal urine concentration and suggest that GSK3α might be one of the initial targets of Li(+) in LiCl-induced nephrogenic diabetes insipidus.
Collapse
Affiliation(s)
- Rikke Nørregaard
- Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Shixin Tao
- The Kidney Institute, Department of Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Line Nilsson
- Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - James R Woodgett
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital and Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada; and
| | - Vijayakumar Kakade
- The Kidney Institute, Department of Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Alan S L Yu
- The Kidney Institute, Department of Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Christiana Howard
- The Kidney Institute, Department of Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Reena Rao
- The Kidney Institute, Department of Medicine, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
36
|
Ahmad F, Lal H, Zhou J, Vagnozzi RJ, Yu JE, Shang X, Woodgett JR, Gao E, Force T. Cardiomyocyte-specific deletion of Gsk3α mitigates post-myocardial infarction remodeling, contractile dysfunction, and heart failure. J Am Coll Cardiol 2014; 64:696-706. [PMID: 25125302 DOI: 10.1016/j.jacc.2014.04.068] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 04/25/2014] [Accepted: 04/30/2014] [Indexed: 01/15/2023]
Abstract
BACKGROUND Injury due to myocardial infarction (MI) is largely irreversible. Once an infarct has occurred, the clinical goal becomes limiting remodeling, preserving left ventricular function, and preventing heart failure. Although traditional approaches (e.g., β-blockers) partially preserve left ventricular function, novel strategies are needed to limit ventricular remodeling post-MI. OBJECTIVES The aim of this study was to determine the role of glycogen synthase kinase-3α (GSK-3α) in post-MI remodeling. METHODS Mice with cardiomyocyte-specific conditional deletion of Gsk3α and littermate controls underwent sham or MI surgery. Heart function was assessed using serial M-mode echocardiography. RESULTS Gsk3α deletion in the heart markedly limits remodeling and preserves left ventricular function post-MI. This is due at least in part to dramatic thinning and expansion of the scar in the control hearts, which was less in the heart of knockout (KO) mice. In contrast, the border zone in the KO mice demonstrated a much thicker scar, and there were more viable cardiomyocytes within the scar/border zone. This was associated with less apoptosis and more proliferation of cardiomyocytes in the KO mice. Mechanistically, reduced apoptosis was due, at least in part, to a marked decrease in the Bax/Bcl-2 ratio, and increased cardiomyocyte proliferation was mediated through cyclin E1 and E2F-1 in the hearts of the KO mice. CONCLUSIONS Taken together, these findings show that reducing GSK-3α expression in cardiomyocytes limits ventricular remodeling and preserves cardiac function post-MI. Specifically targeting GSK-3α could be a novel strategy to limit adverse remodeling and heart failure.
Collapse
Affiliation(s)
- Firdos Ahmad
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA, USA.,Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN,USA
| | - Hind Lal
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA, USA.,Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN,USA
| | - Jibin Zhou
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | - Ronald J Vagnozzi
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | - Justine E Yu
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | - Xiying Shang
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | - James R Woodgett
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Erhe Gao
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | - Thomas Force
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA, USA.,Cardiology Division, Temple University School of Medicine, Philadelphia, PA, USA.,Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN,USA
| |
Collapse
|
37
|
Wang J, Gao E, Chan TO, Zhang XQ, Song J, Shang X, Koch WJ, Feldman AM, Cheung JY. Induced overexpression of Na(+)/Ca(2+) exchanger does not aggravate myocardial dysfunction induced by transverse aortic constriction. J Card Fail 2013; 19:60-70. [PMID: 23273595 DOI: 10.1016/j.cardfail.2012.11.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Revised: 11/06/2012] [Accepted: 11/08/2012] [Indexed: 11/30/2022]
Abstract
BACKGROUND Alterations in expression and activity of cardiac Na(+)/Ca(2+) exchanger (NCX1) have been implicated in the pathogenesis of heart failure. METHODS AND RESULTS Using transgenic mice in which expression of rat NCX1 was induced at 5 weeks of age, we performed transverse aortic constriction (TAC) at 8 weeks and examined cardiac and myocyte function at 15-18 weeks after TAC (age 23-26 weeks). TAC induced left ventricular (LV) and myocyte hypertrophy and increased myocardial fibrosis in both wild-type (WT) and NCX1-overexpressed mice. NCX1 and phosphorylated ryanodine receptor expression was increased by TAC, whereas sarco(endo)plasmic reticulum Ca(2+)-ATPase levels were decreased by TAC. Action potential duration was prolonged by TAC, but to a greater extent in NCX1 myocytes. Na(+)/Ca(2+) exchange current was similar between WT-TAC and WT-sham myocytes, but was higher in NCX1-TAC myocytes. Both myocyte contraction and [Ca(2+)](i) transient amplitudes were reduced in WT-TAC myocytes, but restored to WT-sham levels in NCX1-TAC myocytes. Despite improvement in single myocyte contractility and Ca(2+) dynamics, induced NCX1 overexpression in TAC animals did not ameliorate LV hypertrophy, increase ejection fraction, or enhance inotropic (maximal first derivative of LV pressure rise, +dP/dt) responses to isoproterenol. CONCLUSIONS In pressure-overload hypertrophy, induced overexpression of NCX1 corrected myocyte contractile and [Ca(2+)](i) transient abnormalities but did not aggravate or improve myocardial dysfunction.
Collapse
Affiliation(s)
- Jufang Wang
- Center of Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania 19140, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Zhou J, Freeman TA, Ahmad F, Shang X, Mangano E, Gao E, Farber J, Wang Y, Ma XL, Woodgett J, Vagnozzi RJ, Lal H, Force T. GSK-3α is a central regulator of age-related pathologies in mice. J Clin Invest 2013; 123:1821-32. [PMID: 23549082 DOI: 10.1172/jci64398] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Accepted: 01/24/2013] [Indexed: 12/12/2022] Open
Abstract
Aging is regulated by conserved signaling pathways. The glycogen synthase kinase-3 (GSK-3) family of serine/threonine kinases regulates several of these pathways, but the role of GSK-3 in aging is unknown. Herein, we demonstrate premature death and acceleration of age-related pathologies in the Gsk3a global KO mouse. KO mice developed cardiac hypertrophy and contractile dysfunction as well as sarcomere disruption and striking sarcopenia in cardiac and skeletal muscle, a classical finding in aging. We also observed severe vacuolar degeneration of myofibers and large tubular aggregates in skeletal muscle, consistent with impaired clearance of insoluble cellular debris. Other organ systems, including gut, liver, and the skeletal system, also demonstrated age-related pathologies. Mechanistically, we found marked activation of mTORC1 and associated suppression of autophagy markers in KO mice. Loss of GSK-3α, either by pharmacologic inhibition or Gsk3a gene deletion, suppressed autophagy in fibroblasts. mTOR inhibition rescued this effect and reversed the established pathologies in the striated muscle of the KO mouse. Thus, GSK-3α is a critical regulator of mTORC1, autophagy, and aging. In its absence, aging/senescence is accelerated in multiple tissues. Strategies to maintain GSK-3α activity and/or inhibit mTOR in the elderly could retard the appearance of age-related pathologies.
Collapse
Affiliation(s)
- Jibin Zhou
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania 19140, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Gao E, Koch WJ. A novel and efficient model of coronary artery ligation in the mouse. Methods Mol Biol 2013; 1037:299-311. [PMID: 24029943 DOI: 10.1007/978-1-62703-505-7_17] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Coronary artery ligation to induce myocardial infarction (MI) and ischemia/reperfusion (I/R) injury in mice is typically performed by an invasive and time-consuming approach that requires ventilation and a full thoracotomy (classical method), often resulting in extensive tissue damage and high mortality. Here, we describe a novel and rapid surgical method to induce MI that does not require ventilation. This method is much more efficient and safer than the classical method of MI and I/R injury.
Collapse
Affiliation(s)
- Erhe Gao
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | | |
Collapse
|
40
|
Abstract
Glycogen Synthase Kinase 3 (GSK-3) is a key player in development, physiology and disease. Because of this, GSK-3 inhibitors are increasingly being explored for a variety of applications. In addition most analyses focus on GSK-3β and overlook the closely related protein GSK-3α. Here, we describe novel GSK-3α and GSK-3β mouse alleles that allow us to visualise expression of their respective mRNAs by tracking β-galactosidase activity. We used these new lacZ alleles to compare expression in the palate and cranial sutures and found that there was indeed differential expression. Furthermore, both are loss of function alleles and can be used to generate homozygous mutant mice; in addition, excision of the lacZ cassette from GSK-3α creates a Cre-dependent tissue-specific knockout. As expected, GSK3α mutants were viable, while GSK3β mutants died after birth with a complete cleft palate. We also assessed the GSK-3α mutants for cranial and sternal phenotypes and found that they were essentially normal. Finally, we observed gestational lethality in compound GSK-3β−/−; GSK3α+/− mutants, suggesting that GSK-3 dosage is critical during embryonic development.
Collapse
Affiliation(s)
- William B. Barrell
- Department of Craniofacial Development and Stem Cell Biology, King’s College London, London, United Kingdom
| | - Heather L. Szabo-Rogers
- Department of Craniofacial Development and Stem Cell Biology, King’s College London, London, United Kingdom
| | - Karen J. Liu
- Department of Craniofacial Development and Stem Cell Biology, King’s College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
41
|
Koga K, Kenessey A, Ojamaa K. Macrophage migration inhibitory factor antagonizes pressure overload-induced cardiac hypertrophy. Am J Physiol Heart Circ Physiol 2012; 304:H282-93. [PMID: 23144312 DOI: 10.1152/ajpheart.00595.2012] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Macrophage migration inhibitory factor (MIF) functions as a proinflammatory cytokine when secreted from the cell, but it also exhibits antioxidant properties by virtue of its intrinsic oxidoreductase activity. Since increased production of ROS is implicated in the development of left ventricular hypertrophy, we hypothesized that the redox activity of MIF protects the myocardium when exposed to hemodynamic stress. In a mouse model of myocardial hypertrophy induced by transverse aortic coarctation (TAC) for 10 days, we showed that growth of the MIF-deficient heart was significantly greater by 32% compared with wild-type (WT) TAC hearts and that fibrosis was increased by fourfold (2.62 ± 0.2% vs. 0.6 ± 0.1%). Circulating MIF was increased in TAC animals, and expression of MIF receptor, CD74, was increased in the hypertrophic myocardium. Gene expression analysis showed a 10-fold increase (P < 0.01) in ROS-generating mitochondrial NADPH oxidase and 2- to 3-fold reductions (P < 0.01) in mitochondrial SOD2 and mitochondrial aconitase activities, indicating enhanced oxidative injury in the hypertrophied MIF-deficient ventricle. Hypertrophic signaling pathways showed that phosphorylation of cytosolic glycogen synthase kinase-3α was greater (P < 0.05) at baseline in MIF-deficient hearts than in WT hearts and remained elevated after 10-day TAC. In the hemodynamically stressed MIF-deficient heart, nuclear p21(CIP1) increased sevenfold (P < 0.01), and the cytosolic increase of phospho-p21(CIP1) was significantly greater than in WT TAC hearts. We conclude that MIF antagonizes myocardial hypertrophy and fibrosis in response to hemodynamic stress by maintaining a redox homeostatic phenotype and attenuating stress-induced activation of hypertrophic signaling pathways.
Collapse
Affiliation(s)
- Kiyokazu Koga
- Center for Heart and Lung Research, The Feinstein Institute for Medical Research, Manhasset, NY 11030, USA
| | | | | |
Collapse
|
42
|
Maejima Y, Galeotti J, Molkentin JD, Sadoshima J, Zhai P. Constitutively active MEK1 rescues cardiac dysfunction caused by overexpressed GSK-3α during aging and hemodynamic pressure overload. Am J Physiol Heart Circ Physiol 2012; 303:H979-88. [PMID: 22904158 DOI: 10.1152/ajpheart.00415.2012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Expression of GSK-3α is increased in aging hearts and those subjected to hemodynamic overload. Overexpressed GSK-3α inhibits ERK and enhances pressure overload (PO)-induced cardiac dysfunction. We studied whether suppression of the MEK1/ERK pathway contributes to cardiac responses induced by overexpressed GSK-3α using constitutively active MEK1 (CA-MEK1)/GSK-3α bigenic mice (bigenic mice), which were obtained by crossing cardiac-specific GSK-3α transgenic mice (Tg-GSK) and cardiac-specific CA-MEK1 transgenic mice (Tg-MEK1). The suppression of ERK phosphorylation observed in Tg-GSK was eliminated in bigenic mice. At 12 mo, left ventricular (LV) weight/tibia length, LV weight/body weight, and cardiac myocyte size were significantly smaller in Tg-GSK than in nontransgenic mice (NTg), but were not significantly different between Tg-MEK1 and bigenic mice. The LV ejection fraction (LVEF), fractional shortening (FS), and change in pressure over time were significantly lower in Tg-GSK than in NTg, but were not significantly different between bigenic mice and Tg-MEK1. The increase in apoptosis in Tg-GSK was abolished in bigenic mice, although the increase in fibrosis was not. After PO, the decrease in cardiac hypertrophy and the enhancement of apoptosis seen in Tg-GSK were abrogated in bigenic mice. After PO, the LVEF and FS were significantly reduced in Tg-GSK compared with its sham, but not in NTg, Tg-MEK1, or bigenic mice compared with their respective shams. There was no significant difference in LVEF and FS between bigenic mice and Tg-MEK1 after PO. In conclusion, inhibition of the MEK1/ERK pathway mediates the hypertrophy suppression and cardiac dysfunction caused by GSK-3α overexpression in cardiac myocytes.
Collapse
Affiliation(s)
- Yasuhiro Maejima
- Cardiovascular Research Institute, Department of Cell Biology and Molecular Medicine, University of Medicine and Dentistry, New Jersey, New Jersey Medical School, Newark, New Jersey 07103, USA
| | | | | | | | | |
Collapse
|
43
|
Lo Monte F, Kramer T, Gu J, Anumala UR, Marinelli L, La Pietra V, Novellino E, Franco B, Demedts D, Van Leuven F, Fuertes A, Dominguez JM, Plotkin B, Eldar-Finkelman H, Schmidt B. Identification of Glycogen Synthase Kinase-3 Inhibitors with a Selective Sting for Glycogen Synthase Kinase-3α. J Med Chem 2012; 55:4407-24. [DOI: 10.1021/jm300309a] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Fabio Lo Monte
- Clemens Schöpf—Institute of Organic Chemistry and Biochemistry,
Technische Universität Darmstadt, 64287 Darmstadt, Germany
| | - Thomas Kramer
- Clemens Schöpf—Institute of Organic Chemistry and Biochemistry,
Technische Universität Darmstadt, 64287 Darmstadt, Germany
| | - Jiamin Gu
- Clemens Schöpf—Institute of Organic Chemistry and Biochemistry,
Technische Universität Darmstadt, 64287 Darmstadt, Germany
| | - Upendra Rao Anumala
- Clemens Schöpf—Institute of Organic Chemistry and Biochemistry,
Technische Universität Darmstadt, 64287 Darmstadt, Germany
| | - Luciana Marinelli
- Dipartimento di Chimica Farmaceutica
e Tossicologica, Università di Napoli “Federico II”,
80131 Napoli, Italy
| | - Valeria La Pietra
- Dipartimento di Chimica Farmaceutica
e Tossicologica, Università di Napoli “Federico II”,
80131 Napoli, Italy
| | - Ettore Novellino
- Dipartimento di Chimica Farmaceutica
e Tossicologica, Università di Napoli “Federico II”,
80131 Napoli, Italy
| | - Bénédicte Franco
- Experimental
Genetics Group, Department of Human Genetics, Katholieke Universiteit
Leuven, 3000 Leuven, Belgium
| | - David Demedts
- Experimental
Genetics Group, Department of Human Genetics, Katholieke Universiteit
Leuven, 3000 Leuven, Belgium
| | - Fred Van Leuven
- Experimental
Genetics Group, Department of Human Genetics, Katholieke Universiteit
Leuven, 3000 Leuven, Belgium
| | - Ana Fuertes
- Noscira SA, Drug Discovery, Tres Cantos 28760-Madrid, Spain
| | | | - Batya Plotkin
- Department of Human Molecular Genetics
and Biochemistry, Sackler School of Medicine, Tel Aviv University,
69978 Tel Aviv, Israel
| | - Hagit Eldar-Finkelman
- Department of Human Molecular Genetics
and Biochemistry, Sackler School of Medicine, Tel Aviv University,
69978 Tel Aviv, Israel
| | - Boris Schmidt
- Clemens Schöpf—Institute of Organic Chemistry and Biochemistry,
Technische Universität Darmstadt, 64287 Darmstadt, Germany
| |
Collapse
|
44
|
Lal H, Zhou J, Ahmad F, Zaka R, Vagnozzi RJ, Decaul M, Woodgett J, Gao E, Force T. Glycogen synthase kinase-3α limits ischemic injury, cardiac rupture, post-myocardial infarction remodeling and death. Circulation 2011; 125:65-75. [PMID: 22086876 DOI: 10.1161/circulationaha.111.050666] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND The molecular pathways that regulate the extent of ischemic injury and post-myocardial infarction (MI) remodeling are not well understood. We recently demonstrated that glycogen synthase kinase-3α (GSK-3α) is critical to the heart's response to pressure overload. However, the role, if any, of GSK-3α in regulating ischemic injury and its consequences is not known. METHODS AND RESULTS MI was induced in wild-type (WT) versus GSK-3α((-/-)) (KO) littermates by left anterior descending coronary artery ligation. Pre-MI, WT, and KO hearts had comparable chamber dimensions and ventricular function, but as early as 1 week post-MI, KO mice had significantly more left ventricular dilatation and dysfunction than WT mice. KO mice also had increased mortality during the first 10 days post-MI (43% versus 22%; P=0.04), and postmortem examination confirmed cardiac rupture as the cause of most of the deaths. In the mice that survived the first 10 days, left ventricular dilatation and dysfunction remained worse in the KO mice throughout the study (8 weeks). Hypertrophy, fibrosis, and heart failure were all increased in the KO mice. Given the early deaths due to rupture and the significant reduction in left ventricular function evident as early as 1 week post-MI, we examined infarct size following a 48-hour coronary artery ligation and found it to be increased in the KO mice. This was accompanied by increased apoptosis in the border zone of the MI. This increased susceptibility to ischemic injury-induced apoptosis was also seen in cardiomyocytes isolated from the KO mice that were exposed to hypoxia. Finally, Bax translocation to the mitochondria and cytochrome C release into the cytosol were increased in the KO mice. CONCLUSION GSK-3α confers resistance to ischemic injury, at least in part, via limiting apoptosis. Loss of GSK-3α promotes ischemic injury, increases risk of cardiac rupture, accentuates post-MI remodeling and left ventricular dysfunction, and increases the progression to heart failure. These findings are in striking contrast to multiple previous reports in which deletion or inhibition of GSK-3β is protective.
Collapse
Affiliation(s)
- Hind Lal
- Center for Translational Medicine, Thomas Jefferson University, College Building, Rm 316, 1025 Walnut St., Philadelphia, PA 19107, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Das S, Steenbergen C. Mitochondrial adenine nucleotide transport and cardioprotection. J Mol Cell Cardiol 2011; 52:448-53. [PMID: 21945520 DOI: 10.1016/j.yjmcc.2011.09.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Revised: 09/06/2011] [Accepted: 09/10/2011] [Indexed: 10/17/2022]
Abstract
Mitochondria are highly metabolically active cell organelles that not only act as the powerhouse of the cell by supplying energy through ATP production, but also play a destructive role by initiating cell death pathways. Growing evidence recognizes that mitochondrial dysfunction is one of the major causes of cardiovascular disease. Under de-energized conditions, slowing of adenine nucleotide transport in and out of the mitochondria significantly attenuates myocardial ischemia-reperfusion injury. The purpose of this review is to elaborate on and update the mechanistic pathways which may explain how altered adenine nucleotide transport can influence cardiovascular function. This article is part of a Special Issue entitled "Local Signaling in Myocytes".
Collapse
Affiliation(s)
- Samarjit Das
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | | |
Collapse
|
46
|
Siraskar B, Völkl J, Ahmed MSE, Hierlmeier M, Gu S, Schmid E, Leibrock C, Föller M, Lang UE, Lang F. Enhanced catecholamine release in mice expressing PKB/SGK-resistant GSK3. Pflugers Arch 2011; 462:811-9. [DOI: 10.1007/s00424-011-1006-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Revised: 07/26/2011] [Accepted: 07/28/2011] [Indexed: 12/13/2022]
|
47
|
Inhibition of glycogen synthase kinase 3[beta] activity with lithium in vitro attenuates sepsis-induced changes in muscle protein turnover. Shock 2011; 35:266-74. [PMID: 20926980 DOI: 10.1097/shk.0b013e3181fd068c] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Loss of lean body mass is a characteristic feature of the septic response, and the mechanisms responsible for this decrease and means of prevention have not been fully elucidated. The present study tested the hypothesis that in vitro treatment of skeletal muscle with lithium chloride (LiCl), a glycogen synthase kinase (GSK) 3 inhibitor, would reverse both the sepsis-induced increase in muscle protein degradation and inhibition of protein synthesis. Sepsis decreased GSK-3[beta] phosphorylation and increased GSK-3[beta] activity, under basal conditions. Sepsis increased muscle protein degradation, with a concomitant increase in atrogin 1 and MuRF1 mRNA and 26S proteosome activity. Incubation of septic muscle with LiCl completely reversed the increased GSK-3[beta] activity and decreased proteolysis to basal nonseptic values, but only partially reduced proteosome activity and did not diminish atrogene expression. Lithium chloride also did not ameliorate the sepsis-induced increase in LC3-II, a marker for activated autophagy. In contrast, LiCl increased protein synthesis only in nonseptic control muscle. The inability of septic muscle to respond to LiCl was independent of its ability to reverse the sepsis-induced increase in eukaryotic initiation factor (eIF) 2B[varepsilon] phosphorylation, decreased eIF2B activity, or the reduced phosphorylation of FOXO3, but instead was more closely associated with the continued suppression of mTOR (mammalian target of rapamycin) kinase activity (e.g., reduced phosphorylation of 4E-BP1 and S6). These data suggest that in vitro lithium treatment, which inhibited GSK-3[beta] activity, (a) effectively reversed the sepsis-induced increase in proteolysis, but only in part by a reduction in the ubiquitin-proteosome pathway and not by a reduction in autophagy; and (b) was ineffective at reversing the sepsis-induced decrease in muscle protein synthesis. This lithium-resistant state seems mediated at the level of mTOR and not eIF2/eIF2B. Hence, use of GSK-3[beta] inhibitors in the treatment of sepsis may not be expected to fully correct the imbalance in muscle protein turnover.
Collapse
|
48
|
Tapping the brake on cardiac growth-endogenous repressors of hypertrophic signaling. J Mol Cell Cardiol 2011; 51:156-67. [PMID: 21586293 DOI: 10.1016/j.yjmcc.2011.04.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Revised: 04/26/2011] [Accepted: 04/30/2011] [Indexed: 12/14/2022]
Abstract
Cardiac hypertrophy is considered an early hallmark during the clinical course of heart failure and an important risk factor for cardiac morbidity and mortality. Although hypertrophy of individual cardiomyocytes in response to pathological stimuli has traditionally been considered as an adaptive response required to sustain cardiac output, accumulating evidence from studies in patients and animal models suggests that in most instances hypertrophy of the heart also harbors maladaptive aspects. Major strides have been made in our understanding of the pathways that convey pro-hypertrophic signals from the outside of the cell to the nucleus. In recent years it also has become increasingly evident that the heart possesses a variety of endogenous feedback mechanisms to counterbalance this growth response. These repressive mechanisms are of particular interest since they may provide valuable therapeutic options. In this review we summarize currently known endogenous repressors of pathological cardiac growth as they have been studied by gene targeting in mice. Many of the repressors that function in signal transduction appear to regulate calcineurin (e.g. PICOT, calsarcin, RCAN) and JNK signaling (e.g. CDC42, MKP-1) and some will be described in greater detail in this review. In addition, we will focus on factors such as Kruppel-like factors (KLF4, KLF15 and KLF10) and histone deacetylases (HDACs), which constitute a relevant group of nuclear proteins that repress transcription of the hypertrophic gene program in cardiomyocytes.
Collapse
|
49
|
Cardiotoxicity of kinase inhibitors: the prediction and translation of preclinical models to clinical outcomes. Nat Rev Drug Discov 2011; 10:111-26. [PMID: 21283106 DOI: 10.1038/nrd3252] [Citation(s) in RCA: 258] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
50
|
Cheng H, Woodgett J, Maamari M, Force T. Targeting GSK-3 family members in the heart: a very sharp double-edged sword. J Mol Cell Cardiol 2010; 51:607-13. [PMID: 21163265 DOI: 10.1016/j.yjmcc.2010.11.020] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Revised: 11/26/2010] [Accepted: 11/30/2010] [Indexed: 12/13/2022]
Abstract
The GSK-3 family of serine/threonine kinases, which is comprised of two isoforms (α and β), was initially identified as a negative regulator of glycogen synthase, the rate limiting enzyme of glycogen synthesis [1,2]. In the 30 years since its initial discovery, the family has been reported to regulate a host of additional cellular processes and, consequently, disease states such as bipolar disorders, diabetes, inflammatory diseases, cancer, and neurodegenerative diseases including Alzheimer's Disease and Parkinson's Disease [3,4]. As a result, there has been intense interest on the part of the pharmaceutical industry in developing small molecule antagonists of GSK-3. Herein, we will review the roles played by GSK-3s in the heart, focusing primarily on recent studies that have employed global and tissue-specific gene deletion. We will highlight roles in various pathologic processes, including pressure overload and ischemic injury, focusing on some striking isoform-specific effects of the family. Due to space limitations and/or the relatively limited data in gene-targeted mice, we will not be addressing the family's roles in ischemic pre-conditioning or its many interactions with various pro- and anti-apoptotic factors. This article is part of a special issue entitled "Key Signaling Molecules in Hypertrophy and Heart Failure."
Collapse
Affiliation(s)
- Hui Cheng
- Center for Translational Medicine and Cardiology Division, Thomas Jefferson University Hospital, Philadelphia, PA, USA.
| | | | | | | |
Collapse
|