1
|
Ding L, Lu Z, Jiang X, Zhang S, Tian X, Wang Q. Obesity-derived macrophages upregulate TNF-α to induce apoptosis in glial cell via the NF-κB/PHLPP1 axis. Int Immunopharmacol 2024; 141:112962. [PMID: 39197294 DOI: 10.1016/j.intimp.2024.112962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/14/2024] [Accepted: 08/14/2024] [Indexed: 09/01/2024]
Abstract
Macrophages in obese adipose tissue have been shown to damage nerve fibers, however, the mechanism underlying how macrophages cause glial cell damage remains unknown. This study aimed to characterize the mechanism by which macrophages induce apoptosis in glial cell during obesity formation in mice by single-nucleus RNA sequencing (snRNA-seq). Cells obtained from paraepididymal adipose tissue in obese mice underwent snRNA-seq. Eighteen different clusters were identified, and 12 cell types were annotated, including glial cells, macrophages, and fibroblasts. There was a negative correlation between the number of glial cells and macrophages in mouse adipose tissue during the formation of obesity. The pro-apoptotic factor PHLPP1 was identified in GO Terms. The interaction between adipose tissue glial cells and macrophages was revealed via in-depth analysis, and the cell-cell communication mechanism between the TNF-α and NF-KB/PHLPP1 axes was perfected. Apoptosis of glial cell by upregulation of TNF-α via obesity-derived macrophages and activation of the NF-κB/PHLPP1 axis. We further revealed how macrophages induce apoptosis in glial cells during obesity formation, as well as different changes in the two cell populations. This study provides valuable resources and foundations for understanding the mechanistic effects of macrophages and glial cells during obesity formation, as well as diseases and potential interventions.
Collapse
Affiliation(s)
- Ling Ding
- College of Sport and Health, Shandong Sport University, Jinan, Shandong Province 250102, China
| | - Zhimin Lu
- College of Sport and Health, Shandong Sport University, Jinan, Shandong Province 250102, China
| | - Xing Jiang
- College of Sport and Health, Shandong Sport University, Jinan, Shandong Province 250102, China
| | - Sen Zhang
- College of Sport and Health, Shandong Sport University, Jinan, Shandong Province 250102, China
| | - Xuewen Tian
- College of Sport and Health, Shandong Sport University, Jinan, Shandong Province 250102, China.
| | - Qinglu Wang
- College of Sport and Health, Shandong Sport University, Jinan, Shandong Province 250102, China.
| |
Collapse
|
2
|
Wang Z, Zhang G, Hu S, Fu M, Zhang P, Zhang K, Hao L, Chen S. Research progress on the protective effect of hormones and hormone drugs in myocardial ischemia-reperfusion injury. Biomed Pharmacother 2024; 176:116764. [PMID: 38805965 DOI: 10.1016/j.biopha.2024.116764] [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: 02/21/2024] [Revised: 05/05/2024] [Accepted: 05/17/2024] [Indexed: 05/30/2024] Open
Abstract
Ischemic heart disease (IHD) is a condition where the heart muscle does not receive enough blood flow, leading to cardiac dysfunction. Restoring blood flow to the coronary artery is an effective clinical therapy for myocardial ischemia. This strategy helps lower the size of the myocardial infarction and improves the prognosis of patients. Nevertheless, if the disrupted blood flow to the heart muscle is restored within a specific timeframe, it leads to more severe harm to the previously deprived heart tissue. This condition is referred to as myocardial ischemia/reperfusion injury (MIRI). Until now, there is a dearth of efficacious strategies to prevent and manage MIRI. Hormones are specialized substances that are produced directly into the circulation by endocrine organs or tissues in humans and animals, and they have particular effects on the body. Hormonal medications utilize human or animal hormones as their active components, encompassing sex hormones, adrenaline medications, thyroid hormone medications, and others. While several studies have examined the preventive properties of different endocrine hormones, such as estrogen and hormone analogs, on myocardial injury caused by ischemia-reperfusion, there are other hormone analogs whose mechanisms of action remain unexplained and whose safety cannot be assured. The current study is on hormones and hormone medications, elucidating the mechanism of hormone pharmaceuticals and emphasizing the cardioprotective effects of different endocrine hormones. It aims to provide guidance for the therapeutic use of drugs and offer direction for the examination of MIRI in clinical therapy.
Collapse
Affiliation(s)
- Zhongyi Wang
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Gaojiang Zhang
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Shan Hu
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Meilin Fu
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Pingyuan Zhang
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Kuo Zhang
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Liying Hao
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China.
| | - Sichong Chen
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
3
|
Balamurugan K, Chandra K, Sai Latha S, Swathi M, Joshi MB, Misra P, Parsa KVL. PHLPPs: Emerging players in metabolic disorders. Drug Discov Today 2022; 27:103317. [PMID: 35835313 DOI: 10.1016/j.drudis.2022.07.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/21/2022] [Accepted: 07/07/2022] [Indexed: 12/15/2022]
Abstract
That reversible protein phosphorylation by kinases and phosphatases occurs in metabolic disorders is well known. Various studies have revealed that a multi-faceted and tightly regulated phosphatase, pleckstrin homology domain leucine-rich repeat protein phosphatase (PHLPP)-1/2 displays robust effects in cardioprotection, ischaemia/reperfusion (I/R), and vascular remodelling. PHLPP1 promotes foamy macrophage development through ChREBP/AMPK-dependent pathways. Adipocyte-specific loss of PHLPP2 reduces adiposity, improves glucose tolerance,and attenuates fatty liver via the PHLPP2-HSL-PPARα axis. Discoveries of PHLPP1-mediated insulin resistance and pancreatic β cell death via the PHLPP1/2-Mst1-mTORC1 triangular loop have shed light on its significance in diabetology. PHLPP1 downregulation attenuates diabetic cardiomyopathy (DCM) by restoring PI3K-Akt-mTOR signalling. In this review, we summarise the functional role of, and cellular signalling mediated by, PHLPPs in metabolic tissues and discuss their potential as therapeutic targets.
Collapse
Affiliation(s)
- Keerthana Balamurugan
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences (DRILS), University of Hyderabad Campus, Hyderabad 500046, Telangana, India; Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India
| | - Kanika Chandra
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences (DRILS), University of Hyderabad Campus, Hyderabad 500046, Telangana, India; Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India
| | - S Sai Latha
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences (DRILS), University of Hyderabad Campus, Hyderabad 500046, Telangana, India
| | - M Swathi
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences (DRILS), University of Hyderabad Campus, Hyderabad 500046, Telangana, India
| | - Manjunath B Joshi
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India
| | - Parimal Misra
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences (DRILS), University of Hyderabad Campus, Hyderabad 500046, Telangana, India
| | - Kishore V L Parsa
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences (DRILS), University of Hyderabad Campus, Hyderabad 500046, Telangana, India.
| |
Collapse
|
4
|
Yadav Y, Dey CS. Ser/Thr phosphatases: One of the key regulators of insulin signaling. Rev Endocr Metab Disord 2022; 23:905-917. [PMID: 35697962 DOI: 10.1007/s11154-022-09727-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/18/2022] [Indexed: 10/18/2022]
Abstract
Protein phosphorylation is an important post-translational modification that regulates several cellular processes including insulin signaling. The evidences so far have already portrayed the importance of balanced actions of kinases and phosphatases in regulating the insulin signaling cascade. Therefore, elucidating the role of both kinases and phosphatases are equally important. Unfortunately, the role of phosphatases is less studied as compared to kinases. Since brain responds to insulin and insulin signaling is reported to be crucial for many neuronal processes, it is important to understand the role of neuronal insulin signaling regulators. Ser/Thr phosphatases seem to play significant roles in regulating neuronal insulin signaling. Therefore, in this review, we discussed the involvement of Ser/Thr phosphatases in regulating insulin signaling and insulin resistance in neuronal system at the backdrop of the same phosphatases in peripheral insulin sensitive tissues.
Collapse
Affiliation(s)
- Yamini Yadav
- Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi, New Delhi, 110016, India
| | - Chinmoy Sankar Dey
- Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi, New Delhi, 110016, India.
| |
Collapse
|
5
|
Støle TP, Lunde M, Shen X, Martinsen M, Lunde PK, Li J, Lockwood F, Sjaastad I, Louch WE, Aronsen JM, Christensen G, Carlson CR. The female syndecan-4−/− heart has smaller cardiomyocytes, augmented insulin/pSer473-Akt/pSer9-GSK-3β signaling, and lowered SCOP, pThr308-Akt/Akt and GLUT4 levels. Front Cell Dev Biol 2022; 10:908126. [PMID: 36092718 PMCID: PMC9452846 DOI: 10.3389/fcell.2022.908126] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 07/06/2022] [Indexed: 11/13/2022] Open
Abstract
Background: In cardiac muscle, the ubiquitously expressed proteoglycan syndecan-4 is involved in the hypertrophic response to pressure overload. Protein kinase Akt signaling, which is known to regulate hypertrophy, has been found to be reduced in the cardiac muscle of exercised male syndecan-4−/− mice. In contrast, we have recently found that pSer473-Akt signaling is elevated in the skeletal muscle (tibialis anterior, TA) of female syndecan-4−/− mice. To determine if the differences seen in Akt signaling are sex specific, we have presently investigated Akt signaling in the cardiac muscle of sedentary and exercised female syndecan-4−/− mice. To get deeper insight into the female syndecan-4−/− heart, alterations in cardiomyocyte size, a wide variety of different extracellular matrix components, well-known syndecan-4 binding partners and associated signaling pathways have also been investigated.Methods: Left ventricles (LVs) from sedentary and exercise trained female syndecan-4−/− and WT mice were analyzed by immunoblotting and real-time PCR. Cardiomyocyte size and phosphorylated Ser473-Akt were analyzed in isolated adult cardiomyocytes from female syndecan-4−/− and WT mice by confocal imaging. LV and skeletal muscle (TA) from sedentary male syndecan-4−/− and WT mice were immunoblotted with Akt antibodies for comparison. Glucose levels were measured by a glucometer, and fasting blood serum insulin and C-peptide levels were measured by ELISA.Results: Compared to female WT hearts, sedentary female syndecan-4−/− LV cardiomyocytes were smaller and hearts had higher levels of pSer473-Akt and its downstream target pSer9-GSK-3β. The pSer473-Akt inhibitory phosphatase PHLPP1/SCOP was lowered, which may be in response to the elevated serum insulin levels found in the female syndecan-4−/− mice. We also observed lowered levels of pThr308-Akt/Akt and GLUT4 in the female syndecan-4−/− heart and an increased LRP6 level after exercise. Otherwise, few alterations were found. The pThr308-Akt and pSer473-Akt levels were unaltered in the cardiac and skeletal muscles of sedentary male syndecan-4−/− mice.Conclusion: Our data indicate smaller cardiomyocytes, an elevated insulin/pSer473-Akt/pSer9-GSK-3β signaling pathway, and lowered SCOP, pThr308-Akt/Akt and GLUT4 levels in the female syndecan-4−/− heart. In contrast, cardiomyocyte size, and Akt signaling were unaltered in both cardiac and skeletal muscles from male syndecan-4−/− mice, suggesting important sex differences.
Collapse
Affiliation(s)
- Thea Parsberg Støle
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- *Correspondence: Thea Parsberg Støle,
| | - Marianne Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Xin Shen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Marita Martinsen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Per Kristian Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Jia Li
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Francesca Lockwood
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - William Edward Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Jan Magnus Aronsen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Geir Christensen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Cathrine Rein Carlson
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| |
Collapse
|
6
|
Pahlavani HA. Exercise-induced signaling pathways to counteracting cardiac apoptotic processes. Front Cell Dev Biol 2022; 10:950927. [PMID: 36036015 PMCID: PMC9403089 DOI: 10.3389/fcell.2022.950927] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 07/15/2022] [Indexed: 01/15/2023] Open
Abstract
Cardiovascular diseases are the most common cause of death in the world. One of the major causes of cardiac death is excessive apoptosis. However, multiple pathways through moderate exercise can reduce myocardial apoptosis. After moderate exercise, the expression of anti-apoptotic proteins such as IGF-1, IGF-1R, p-PI3K, p-Akt, ERK-1/2, SIRT3, PGC-1α, and Bcl-2 increases in the heart. While apoptotic proteins such as PTEN, PHLPP-1, GSK-3, JNK, P38MAPK, and FOXO are reduced in the heart. Exercise-induced mechanical stress activates the β and α5 integrins and subsequently, focal adhesion kinase phosphorylation activates the Akt/mTORC1 and ERK-1/2 pathways, leading to an anti-apoptotic response. One of the reasons for the decrease in exercise-induced apoptosis is the decrease in Fas-ligand protein, Fas-death receptor, TNF-α receptor, Fas-associated death domain (FADD), caspase-8, and caspase-3. In addition, after exercise mitochondrial-dependent apoptotic factors such as Bid, t-Bid, Bad, p-Bad, Bak, cytochrome c, and caspase-9 are reduced. These changes lead to a reduction in oxidative damage, a reduction in infarct size, a reduction in cardiac apoptosis, and an increase in myocardial function. After exercising in the heart, the levels of RhoA, ROCK1, Rac1, and ROCK2 decrease, while the levels of PKCε, PKCδ, and PKCɑ are activated to regulate calcium and prevent mPTP perforation. Exercise has an anti-apoptotic effect on heart failure by increasing the PKA-Akt-eNOS and FSTL1-USP10-Notch1 pathways, reducing the negative effects of CaMKIIδ, and increasing the calcineurin/NFAT pathway. Exercise plays a protective role in the heart by increasing HSP20, HSP27, HSP40, HSP70, HSP72, and HSP90 along with increasing JAK2 and STAT3 phosphorylation. However, research on exercise and factors such as Pim-1, Notch, and FAK in cardiac apoptosis is scarce, so further research is needed. Future research is recommended to discover more anti-apoptotic pathways. It is also recommended to study the synergistic effect of exercise with gene therapy, dietary supplements, and cell therapy for future research.
Collapse
|
7
|
Lemoine KA, Fassas JM, Ohannesian SH, Purcell NH. On the PHLPPside: Emerging roles of PHLPP phosphatases in the heart. Cell Signal 2021; 86:110097. [PMID: 34320369 PMCID: PMC8403656 DOI: 10.1016/j.cellsig.2021.110097] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 07/20/2021] [Accepted: 07/22/2021] [Indexed: 02/07/2023]
Abstract
PH domain leucine-rich repeat protein phosphatase (PHLPP) is a family of enzymes made up of two isoforms (PHLPP1 and PHLPP2), whose actions modulate intracellular activity via the dephosphorylation of specific serine/threonine (Ser/Thr) residues on proteins such as Akt. Recent data generated in our lab, supported by findings from others, implicates the divergent roles of PHLPP1 and PHLPP2 in maintaining cellular homeostasis since dysregulation of these enzymes has been linked to various pathological states including cardiovascular disease, diabetes, ischemia/reperfusion injury, musculoskeletal disease, and cancer. Therefore, development of therapies to modulate specific isoforms of PHLPP could prove to be therapeutically beneficial in several diseases especially those targeting the cardiovascular system. This review is intended to provide a comprehensive summary of current literature detailing the role of the PHLPP isoforms in the development and progression of heart disease.
Collapse
Affiliation(s)
- Kellie A Lemoine
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92039, USA
| | - Julianna M Fassas
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92039, USA
| | - Shirag H Ohannesian
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92039, USA
| | - Nicole H Purcell
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92039, USA; Cardiovascular Molecular Signaling, Huntington Medical Research Institutes, Pasadena, CA 91105, USA.
| |
Collapse
|
8
|
Therapies to prevent post-infarction remodelling: From repair to regeneration. Biomaterials 2021; 275:120906. [PMID: 34139506 DOI: 10.1016/j.biomaterials.2021.120906] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 05/02/2021] [Accepted: 05/20/2021] [Indexed: 12/15/2022]
Abstract
Myocardial infarction is the first cause of worldwide mortality, with an increasing incidence also reported in developing countries. Over the past decades, preclinical research and clinical trials continually tested the efficacy of cellular and acellular-based treatments. However, none of them resulted in a drug or device currently used in combination with either percutaneous coronary intervention or coronary artery bypass graft. Inflammatory, proliferation and remodelling phases follow the ischaemic event in the myocardial tissue. Only recently, single-cell sequencing analyses provided insights into the specific cell populations which determine the final fibrotic deposition in the affected region. In this review, ischaemia, inflammation, fibrosis, angiogenesis, cellular stress and fundamental cellular and molecular components are evaluated as therapeutic targets. Given the emerging evidence of biomaterial-based systems, the increasing use of injectable hydrogels/scaffolds and epicardial patches is reported both as acellular and cellularised/functionalised treatments. Since several variables influence the outcome of any experimented treatment, we return to the pathological basis with an unbiased view towards any specific process or cellular component. Thus, by evaluating the benefits and limitations of the approaches based on these targets, the reader can weigh the rationale of each of the strategies that reached the clinical trials stage. As recent studies focused on the relevance of the extracellular matrix in modulating ischaemic remodelling and enhancing myocardial regeneration, we aim to portray current trends in the field with this review. Finally, approaches towards feasible translational studies that are as yet unexplored are also suggested.
Collapse
|
9
|
Budiono BP, See Hoe LE, Peart JN, Vider J, Ashton KJ, Jacques A, Haseler LJ, Headrick JP. Effects of voluntary exercise duration on myocardial ischaemic tolerance, kinase signaling and gene expression. Life Sci 2021; 274:119253. [PMID: 33647270 DOI: 10.1016/j.lfs.2021.119253] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 12/20/2022]
Abstract
AIM Exercise is cardioprotective, though optimal interventions are unclear. We assessed duration dependent effects of exercise on myocardial ischemia-reperfusion (I-R) injury, kinase signaling and gene expression. METHODS Responses to brief (2 day; 2EX), intermediate (7 and 14 day; 7EX and 14EX) and extended (28 day; 28EX) voluntary wheel running (VWR) were studied in male C57Bl/6 mice. Cardiac function, I-R tolerance and survival kinase signaling were assessed in perfused hearts. KEY FINDINGS Mice progressively increased running distances and intensity, from 2.4 ± 0.2 km/day (0.55 ± 0.04 m/s) at 2-days to 10.6 ± 0.4 km/day (0.72 ± 0.06 m/s) after 28-days. Myocardial mass and contractility were modified at 14-28 days VWR. Cardioprotection was not 'dose-dependent', with I-R tolerance enhanced within 7 days and not further improved with greater VWR duration, volume or intensity. Protection was associated with AKT, ERK1/2 and GSK3β phosphorylation, with phospho-AMPK selectively enhanced with brief VWR. Gene expression was duration-dependent: 7 day VWR up-regulated glycolytic (Pfkm) and down-regulated maladaptive remodeling (Mmp2) genes; 28 day VWR up-regulated caveolar (Cav3), mitochondrial biogenesis (Ppargc1a, Sirt3) and titin (Ttn) genes. Interestingly, I-R tolerance in 2EX/2SED groups improved vs. groups subjected to longer sedentariness, suggesting transient protection on transition to housing with running wheels. SIGNIFICANCE Cardioprotection is induced with as little as 7 days VWR, yet not enhanced with further or faster running. This protection is linked to survival kinase phospho-regulation (particularly AKT and ERK1/2), with glycolytic, mitochondrial, caveolar and myofibrillar gene changes potentially contributing. Intriguingly, environmental enrichment may also protect via similar kinase regulation.
Collapse
Affiliation(s)
- Boris P Budiono
- Charles Sturt University, School of Community Health, Port Macquarie, NSW, Australia
| | - Louise E See Hoe
- Griffith University, School of Medical Science, Gold Coast, QLD, Australia
| | - Jason N Peart
- Griffith University, School of Medical Science, Gold Coast, QLD, Australia
| | - Jelena Vider
- Griffith University, School of Medical Science, Gold Coast, QLD, Australia
| | - Kevin J Ashton
- Bond University, Faculty of Health and Medicine, Robina, QLD, Australia
| | - Angela Jacques
- Curtin University, School of Physiotherapy and Exercise Science, Bentley, WA, Australia
| | - Luke J Haseler
- Curtin University, School of Physiotherapy and Exercise Science, Bentley, WA, Australia
| | - John P Headrick
- Griffith University, School of Medical Science, Gold Coast, QLD, Australia.
| |
Collapse
|
10
|
Li T, Yin Y, Mu N, Wang Y, Liu M, Chen M, Jiang W, Yu L, Li Y, Ma H. Metformin-Enhanced Cardiac AMP-Activated Protein Kinase/Atrogin-1 Pathways Inhibit Charged Multivesicular Body Protein 2B Accumulation in Ischemia-Reperfusion Injury. Front Cell Dev Biol 2021; 8:621509. [PMID: 33614629 PMCID: PMC7892907 DOI: 10.3389/fcell.2020.621509] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 12/22/2020] [Indexed: 12/19/2022] Open
Abstract
Background: Cardiac autophagic flux is impaired during myocardial ischemia/reperfusion (MI/R). Impaired autophagic flux may exacerbate MI/R injury. Charged multivesicular body protein 2B (CHMP2B) is a subunit of the endosomal sorting complex required for transport (ESCRT-III) complex that is required for autophagy. However, the reverse role of CHMP2B accumulation in autophagy and MI/R injury has not been established. The objective of this article is to elucidate the roles of AMP-activated protein kinase (AMPK)/atrogin-1 pathways in inhibiting CHMP2B accumulation in ischemia-reperfusion injury. Methods: Male C57BL/6 mice (3-4 months) and H9c2 cardiomyocytes were used to evaluate MI/R and hypoxia/reoxygenation (H/R) injury in vivo and in vitro, respectively. MI/R was built by a left lateral thoracotomy and occluded the left anterior descending artery. H9c2 cells were firstly treated in 95% N2 and 5% CO2 for 15 h and reoxygenation for 1 h. Metformin (100 mg/kg/d) and CHMP2B (Ad-CHMP2B) transfected adenoviruses were administered to the mice. The H9c2 cells were treated with metformin (2.5 mM), MG-132 (10 μM), bafilomycin A1 (10 nM), and compound C (20 μM). Results: Autophagic flux was found to be inhibited in H/R-treated cardiomyocytes and MI/R mice, with elevated cardiac CHMP2B accumulation. Upregulated CHMP2B levels in the in vivo and in vitro experiments were shown to inhibit autophagic flux leading to the deterioration of H/R-cardiomyocytes and MI/R injury. This finding implies that CHMP2B accumulation increases the risk of myocardial ischemia. Metformin suppressed CHMP2B accumulation and ameliorated H/R-induced autophagic dysfunction by activating AMPK. Activated AMPK upregulated the messenger RNA expression and protein levels of atrogin-1, a muscle-specific ubiquitin ligase, in the myocardium. Atrogin-1 significantly enhanced the interaction between atrogin-1 and CHMP2B, therefore, promoting CHMP2B degradation in the MI/R myocardium. Finally, this study revealed that metformin-inhibited CHMP2B accumulation induced autophagic impairment and ischemic susceptibility in vivo through the AMPK-regulated CHMP2B degradation by atrogin-1. Conclusion: Impaired CHMP2B clearance in vitro and in vivo inhibits autophagic flux and weakens the myocardial ischemic tolerance. Metformin treatment degrades CHMP2B through the AMPK-atrogin-1-dependent pathway to maintain the homeostasis of autophagic flux. This is a novel mechanism that enriches the understanding of cardioprotection.
Collapse
Affiliation(s)
- Tian Li
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Yue Yin
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Nan Mu
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Yishi Wang
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Manling Liu
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Mai Chen
- Department of Cardiovascular Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Wenhua Jiang
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, China
| | - Lu Yu
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yan Li
- Department of Cardiovascular Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Heng Ma
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
11
|
Blondelle J, Biju A, Lange S. The Role of Cullin-RING Ligases in Striated Muscle Development, Function, and Disease. Int J Mol Sci 2020; 21:E7936. [PMID: 33114658 PMCID: PMC7672578 DOI: 10.3390/ijms21217936] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/11/2020] [Accepted: 10/13/2020] [Indexed: 02/07/2023] Open
Abstract
The well-orchestrated turnover of proteins in cross-striated muscles is one of the fundamental processes required for muscle cell function and survival. Dysfunction of the intricate protein degradation machinery is often associated with development of cardiac and skeletal muscle myopathies. Most muscle proteins are degraded by the ubiquitin-proteasome system (UPS). The UPS involves a number of enzymes, including E3-ligases, which tightly control which protein substrates are marked for degradation by the proteasome. Recent data reveal that E3-ligases of the cullin family play more diverse and crucial roles in cross striated muscles than previously anticipated. This review highlights some of the findings on the multifaceted functions of cullin-RING E3-ligases, their substrate adapters, muscle protein substrates, and regulatory proteins, such as the Cop9 signalosome, for the development of cross striated muscles, and their roles in the etiology of myopathies.
Collapse
Affiliation(s)
- Jordan Blondelle
- Department of Medicine, University of California, La Jolla, CA 92093, USA
| | - Andrea Biju
- Department of Medicine, University of California, La Jolla, CA 92093, USA
| | - Stephan Lange
- Department of Medicine, University of California, La Jolla, CA 92093, USA
- Department of Molecular and Clinical Medicine, University of Gothenburg, 41345 Gothenburg, Sweden
| |
Collapse
|
12
|
Zhang X, Lu Y, He N, Wang F. Downregulation of PHLPP1 ameliorates high glucose-evoked injury in retinal ganglion cells by attenuating apoptosis and oxidative stress through enhancement of Nrf2 activation. Exp Cell Res 2020; 397:112344. [PMID: 33164862 DOI: 10.1016/j.yexcr.2020.112344] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 12/18/2022]
Abstract
High glucose (HG)-induced oxidative stress contributes significantly to the pathogenesis of diabetic retinopathy. Pleckstrin homology domain and leucine rich repeat protein phosphatase 1 (PHLPP1) has emerged as a key regulator of oxidative stress implicated in various pathological processes. However, whether PHLPP1 participates in the regulation of HG-induced oxidative stress injury of retinal ganglion cells (RGCs) in diabetic retinopathy is undetermined. The purpose of this study was to explore the potential role and molecular mechanism of PHLPP1 in regulating HG-induced injury of RGCs. Our data showed that PHLPP1 expression was markedly elevated in RGCs from diabetic rats and HG-exposed RGCs. Our functional assay elucidated that knockdown of PHLPP1 improved cell viability and decreased cell apoptosis and reactive oxygen species (ROS) production in HG-exposed RGCs. Additionally, upregulation of PHLPP1 lowered cell viability and increased cell apoptosis and ROS production in HG-exposed RGCs. Mechanistically, knockdown of PHLPP1 resulted in an increase in nuclear factor erythroid-2 related factor 2 (Nrf2) nuclear expression and Nrf2/antioxidant response element (ARE)-mediated transcription associated with upregulation of glycogen synthase kinase-3β (GSK-3β) phosphorylation. Moreover, inhibition of GSK-3β significantly reversed the suppressive effect of PHLPP1 overexpression on Nrf2/ARE activation. Notably, the protective effect of PHLPP1 knockdown on HG-induced injury in RGCs was markedly abolished by Nrf2 inhibition. In conclusion, Our findings demonstrate that downregulation of PHLPP1 activates Nrf2/ARE signaling to protect RGCs from HG-induced apoptosis and oxidative stress. This study indicates a potential role of PHLPP1 in regulating HG-induced injury of RGCs during the development and progression of diabetic retinopathy.
Collapse
Affiliation(s)
- Xiaohui Zhang
- Ophthalmology Department, The Second Affiliated Hospital, Medical College, Xi'an Jiaotong University, Xi'an, 710004, PR China
| | - Ye Lu
- Ophthalmology Department, The Second Affiliated Hospital, Medical College, Xi'an Jiaotong University, Xi'an, 710004, PR China
| | - Na He
- Ophthalmology Department, The Second Affiliated Hospital, Medical College, Xi'an Jiaotong University, Xi'an, 710004, PR China
| | - Feng Wang
- Ophthalmology Department, The Second Affiliated Hospital, Medical College, Xi'an Jiaotong University, Xi'an, 710004, PR China.
| |
Collapse
|
13
|
Dai G, Li Y, Liu J, Zhang C, Chen M, Lu P, Rui Y. Higher BMP Expression in Tendon Stem/Progenitor Cells Contributes to the Increased Heterotopic Ossification in Achilles Tendon With Aging. Front Cell Dev Biol 2020; 8:570605. [PMID: 33102476 PMCID: PMC7546413 DOI: 10.3389/fcell.2020.570605] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 09/04/2020] [Indexed: 12/18/2022] Open
Abstract
Although the mineralization in tendon tissue has been reported in a series of aging and disease models, the underlying mechanisms remain unknown. This study aimed to describe the appearance of heterotopic ossification in rat Achilles tendon and further verify whether this tissue metaplasia is related to the enhanced osteogenic differentiation of tendon stem/progenitor cells (TSPCs) owing to the higher expression of bone morphogenetic proteins (BMP-2/4/7) with aging. The male SD rats, aged 4, 8, and 20 months (M), were used. The analyses of ossification and BMP expression in tendon were tested by radiological view (X-ray and CT), histological staining [hematoxylin and eosin (HE), Alcian blue, and Alizarin red], immunohistochemistry, and Western blot. The osteogenic differentiation potential and BMP expression of TSPCs were examined by Alizarin red S staining and real-time PCR. TSPCs were treated with BMP-2 or noggin, and the osteogenic differentiation potential was also examined. X-ray and CT showed the appearance of heterotopic ossification in tendon, and the volume and density of ossification was increased with aging. Histological staining showed the appearance of calcified region surrounded by chondrocyte-like cells and the increased osteogenesis-related gene and BMP expression in ossified tendon with aging. Moreover, the osteogenic differentiation potential and BMP expression in TSPCs isolated from ossified tendon were increased with aging. Additionally, BMP-2 increased the calcium nodule formation and osteogenesis-related gene expression in TSPCs. The addition of noggin inhibited BMP-induced enhancement of osteogenic differentiation. Thus, these findings suggested that the enhanced osteogenic differentiation of TSPCs contributes to the increased heterotopic ossification in aged tendon, which might be induced by the higher expression of BMPs with aging.
Collapse
Affiliation(s)
- Guangchun Dai
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- School of Medicine, Southeast University, Nanjing, China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, China
- Trauma Center, Zhongda Hospital, Southeast University, Nanjing, China
| | - Yingjuan Li
- Department of Geriatrics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Junyan Liu
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- School of Medicine, Southeast University, Nanjing, China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, China
- Trauma Center, Zhongda Hospital, Southeast University, Nanjing, China
| | - Cheng Zhang
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- School of Medicine, Southeast University, Nanjing, China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, China
- Trauma Center, Zhongda Hospital, Southeast University, Nanjing, China
| | - Minhao Chen
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- School of Medicine, Southeast University, Nanjing, China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, China
- Trauma Center, Zhongda Hospital, Southeast University, Nanjing, China
| | - Panpan Lu
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- School of Medicine, Southeast University, Nanjing, China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, China
- Trauma Center, Zhongda Hospital, Southeast University, Nanjing, China
| | - Yunfeng Rui
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, China
- Trauma Center, Zhongda Hospital, Southeast University, Nanjing, China
- China Orthopedic Regenerative Medicine Group, Hangzhou, China
| |
Collapse
|
14
|
Zhang M, Wang X, Liu M, Liu D, Pan J, Tian J, Jin T, Xu Y, An F. Inhibition of PHLPP1 ameliorates cardiac dysfunction via activation of the PI3K/Akt/mTOR signalling pathway in diabetic cardiomyopathy. J Cell Mol Med 2020; 24:4612-4623. [PMID: 32150791 PMCID: PMC7176843 DOI: 10.1111/jcmm.15123] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 12/18/2019] [Accepted: 02/11/2020] [Indexed: 12/20/2022] Open
Abstract
Background Pleckstrin homology (PH) domain leucine‐rich repeat protein phosphatase 1 (PHLPP1) is a kind of serine/threonine phosphatase, whose dysregulation is accompanied with numerous human diseases. However, its role in diabetic cardiomyopathy remains unclear. We explored the underlying function and mechanism of PHLPP1 in diabetic cardiomyopathy (DCM). Method In vivo, Type 1 diabetic rats were induced by intraperitoneal injection of 60 mg/kg streptozotocin (STZ). Lentivirus‐mediated short hairpin RNA (shRNA) was used to knock down the expression of PHLPP1. In vitro, primary neonatal rat cardiomyocytes and H9C2 cells were incubated in 5.5 mmol/L glucose (normal glucose, NG) or 33.3 mmol/L glucose (high glucose, HG). PHLPP1 expression was inhibited by PHLPP1‐siRNA to probe into the function of PHLPP1 in high glucose‐induced apoptosis in H9c2 cells. Results Diabetic rats showed up‐regulated PHLPP1 expression, left ventricular dysfunction, increased myocardial apoptosis and fibrosis. PHLPP1 inhibition alleviated cardiac dysfunction. Additionally, PHLPP1 inhibition significantly reduced HG‐induced apoptosis and restored PI3K/AKT/mTOR pathway activity in H9c2 cells. Furthermore, pretreatment with LY294002, an inhibitor of PI3K/Akt/mTOR pathway, abolished the anti‐apoptotic effect of PHLPP1 inhibition. Conclusion Our study indicated that PHLPP1 inhibition alleviated cardiac dysfunction via activating the PI3K/Akt/mTOR signalling pathway in DCM. Therefore, PHLPP1 may be a novel therapeutic target for human DCM.
Collapse
Affiliation(s)
- Mingjun Zhang
- Department of Cardiology, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Xuyang Wang
- Department of Cardiology, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Ming Liu
- Department of Cardiology, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Dian Liu
- Department of Cardiology, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Jinyu Pan
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital of Shandong First Medical University, Jinan, China
| | - Jingjing Tian
- Department of Cardiology, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Tao Jin
- Department of Cardiology, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Yunfan Xu
- College of Life Sciences, Sichuan University, Chengdu, China
| | - Fengshuang An
- Department of Cardiology, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
15
|
Li C, Mu N, Gu C, Liu M, Yang Z, Yin Y, Chen M, Wang Y, Han Y, Yu L, Ma H. Metformin mediates cardioprotection against aging-induced ischemic necroptosis. Aging Cell 2020; 19:e13096. [PMID: 31944526 PMCID: PMC6996959 DOI: 10.1111/acel.13096] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 10/23/2019] [Accepted: 12/08/2019] [Indexed: 12/13/2022] Open
Abstract
Necroptosis is crucially involved in severe cardiac pathological conditions. However, whether necroptosis contributes to age‐related intolerance to ischemia/reperfusion (I/R) injury remains elusive. In addition, metformin as a potential anti‐aging related injury drug, how it interacts with myocardial necroptosis is not yet clear. Male C57BL/6 mice at 3–4‐ (young) and 22–24 months of age (aged) and RIPK3‐deficient (Ripk3−/−) mice were used to investigate aging‐related I/R injury in vivo. Metformin (125 μg/kg, i.p.), necrostatin‐1 (3.5 mg/kg), and adenovirus vector encoding p62‐shRNAs (Ad‐sh‐p62) were used to treat aging mice. I/R‐induced myocardial necroptosis was exaggerated in aged mice, which correlated with autophagy defects characterized by p62 accumulation in aged hearts or aged human myocardium. Functionally, blocking autophagic flux promoted H/R‐evoked cardiomyocyte necroptosis in vitro. We further revealed that p62 forms a complex with RIP1‐RIP3 (necrosome) and promotes the binding of RIP1 and RIP3. In mice, necrostatin‐1 treatment (a RIP1 inhibitor), RIP3 deficiency, and cardiac p62 knockdown in vivo demonstrated that p62‐RIP1‐RIP3‐dependent myocardial necroptosis contributes to aging‐related myocardial vulnerability to I/R injury. Notably, metformin treatment disrupted p62‐RIP1‐RIP3 complexes and effectively repressed I/R‐induced necroptosis in aged hearts, ultimately reducing mortality in this model. These findings highlight previously unknown mechanisms of aging‐related myocardial ischemic vulnerability: p62‐necrosome‐dependent necroptosis. Metformin acts as a cardioprotective agent that inhibits this unfavorable chain mechanism of aging‐related I/R susceptibility.
Collapse
Affiliation(s)
- Chen Li
- Department of Physiology and Pathophysiology Fourth Military Medical University Xi'an China
| | - Nan Mu
- Department of Physiology and Pathophysiology Fourth Military Medical University Xi'an China
| | - Chunhu Gu
- Department of Cardiovascular Surgery Xijing Hospital Fourth Military Medical University Xi'an China
| | - Manling Liu
- Department of Physiology and Pathophysiology Fourth Military Medical University Xi'an China
| | - Zheng Yang
- Department of Physiology and Pathophysiology Fourth Military Medical University Xi'an China
| | - Yue Yin
- Department of Physiology and Pathophysiology Fourth Military Medical University Xi'an China
| | - Mai Chen
- Department of Cardiovascular Medicine Xijing Hospital Fourth Military Medical University Xi'an China
| | - Yishi Wang
- Department of Physiology and Pathophysiology Fourth Military Medical University Xi'an China
| | - Yuehu Han
- Department of Cardiovascular Surgery Xijing Hospital Fourth Military Medical University Xi'an China
| | - Lu Yu
- Department of Pathology Xijing Hospital Fourth Military Medical University Xi'an China
| | - Heng Ma
- Department of Physiology and Pathophysiology Fourth Military Medical University Xi'an China
| |
Collapse
|
16
|
Wang Y, Tian MM, Mi CJ, Chen KL, Ji YC, Wang L, Zhang J, Cheng K. Exercise protects the heart against myocardial infarction through upregulation of miR-1192. Biochem Biophys Res Commun 2020; 521:1061-1069. [DOI: 10.1016/j.bbrc.2019.11.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 11/02/2019] [Indexed: 12/11/2022]
|
17
|
Hou Z, Qin X, Hu Y, Zhang X, Li G, Wu J, Li J, Sha J, Chen J, Xia J, Wang L, Gao F. Longterm Exercise-Derived Exosomal miR-342-5p. Circ Res 2019; 124:1386-1400. [DOI: 10.1161/circresaha.118.314635] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Zuoxu Hou
- From the School of Aerospace Medicine (Z.H., X.Q., Y.H., X.Z., G.L., J.W., J.L., F.G.), Fourth Military Medical University, Xi’an, China
| | - Xinghua Qin
- From the School of Aerospace Medicine (Z.H., X.Q., Y.H., X.Z., G.L., J.W., J.L., F.G.), Fourth Military Medical University, Xi’an, China
| | - Yuanyuan Hu
- From the School of Aerospace Medicine (Z.H., X.Q., Y.H., X.Z., G.L., J.W., J.L., F.G.), Fourth Military Medical University, Xi’an, China
| | - Xing Zhang
- From the School of Aerospace Medicine (Z.H., X.Q., Y.H., X.Z., G.L., J.W., J.L., F.G.), Fourth Military Medical University, Xi’an, China
| | - Guohua Li
- From the School of Aerospace Medicine (Z.H., X.Q., Y.H., X.Z., G.L., J.W., J.L., F.G.), Fourth Military Medical University, Xi’an, China
| | - Jie Wu
- From the School of Aerospace Medicine (Z.H., X.Q., Y.H., X.Z., G.L., J.W., J.L., F.G.), Fourth Military Medical University, Xi’an, China
| | - Jia Li
- From the School of Aerospace Medicine (Z.H., X.Q., Y.H., X.Z., G.L., J.W., J.L., F.G.), Fourth Military Medical University, Xi’an, China
| | - Jianding Sha
- Department of Physical Education (J.S.), Fourth Military Medical University, Xi’an, China
| | - Jiangwei Chen
- From the School of Aerospace Medicine (Z.H., X.Q., Y.H., X.Z., G.L., J.W., J.L., F.G.), Fourth Military Medical University, Xi’an, China
| | - Jielai Xia
- Department of Health Statistics (J.X.), Fourth Military Medical University, Xi’an, China
| | - Lifeng Wang
- Department of Biochemistry and Molecular Biology (L.W.), Fourth Military Medical University, Xi’an, China
| | - Feng Gao
- From the School of Aerospace Medicine (Z.H., X.Q., Y.H., X.Z., G.L., J.W., J.L., F.G.), Fourth Military Medical University, Xi’an, China
| |
Collapse
|
18
|
Huang Q, Liu C, Li JR, Zhang L, Huang FC, Wang D, Luo YJ. Incremental effect of liraglutide on traditional insulin injections in rats with type 2 diabetes mellitus by maintaining glycolipid metabolism and cardiovascular function. Exp Ther Med 2019; 17:1863-1869. [PMID: 30783461 DOI: 10.3892/etm.2019.7148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/26/2018] [Indexed: 11/06/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is characterized by chronic hyperglycemia, damaged insulin secretion and insulin resistance with high morbidity and mortality. Liraglutide (liragl) and insulin are effective hypoglycemic agents used in T2DM treatment. The potential effect of liragl in combination with insulin on T2DM remains unclear. The aim of the current study was to explore effects of liragl combined with insulin on glycolipid metabolism and cardiovascular function in rats with diabetes. A diabetes model was established in Sprague Dawley rats exposed to a high calorie and high sugar diet in conjunction with intraperitoneal injections of streptozotocin. Results indicated that liragl or insulin used alone decreased glucose and elevated insulin and c-peptide levels. However, their combination revealed greater effects. A significant increase in high-density lipoprotein cholesterol levels along with a decrease in total cholesterol, triglycerides and low-density lipoprotein cholesterol were observed in liragl- and insulin-treated rats compared with STZ-induced diabetes rats. Furthermore, co-administration of liragl and insulin significantly decreased sterol regulatory element-binding protein 1 levels and increased adenosine 5'-monophosphate kinase-α1 and carnitine palmitoyltransferase 1 expression. Combining liragl with insulin reduced myocardial hypertrophy level and gaps between cardiomyocytes compared with liragl or insulin treatment alone. Caspase-3 expression was significantly decreased by combination treatment of liragl and insulin. Oxidative damage was significantly decreased by co-administration of liragl and insulin through enhancing superoxide dismutase expression and reducing malondialdehyde. Furthermore, combination of liragl and insulin significantly reduced myocardial enzyme expression, including myoglobin, creatine kinase-muscle/brain and cardiac troponin I. In summary, the current study demonstrated synergistic effects of liragl and insulin injections on a T2DM rat model by maintaining glycolipid metabolism and cardiovascular function.
Collapse
Affiliation(s)
- Qian Huang
- Department of Endocrinology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Chan Liu
- Department of Endocrinology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Jia-Rui Li
- Department of Endocrinology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Ling Zhang
- Department of Endocrinology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Fu-Chang Huang
- Department of Endocrinology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Dan Wang
- Department of Endocrinology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Ya-Jing Luo
- Department of Endocrinology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| |
Collapse
|
19
|
Tang B, Ma J, Ha X, Zhang Y, Xing Y. Tumor necrosis factor-alpha upregulated PHLPP1 through activating nuclear factor-kappa B during myocardial ischemia/reperfusion. Life Sci 2018; 207:355-363. [PMID: 29940243 DOI: 10.1016/j.lfs.2018.06.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 06/19/2018] [Accepted: 06/21/2018] [Indexed: 12/18/2022]
Abstract
AIMS The pleckstrin homology domain leucine-rich repeat protein phosphatase 1 (PHLPP1) specifically regulates phospho-Ser473 of protein kinase B (PKB, Akt) opposing cell survival during myocardial ischemia/reperfusion (I/R). Previous studies demonstrated PHLPP1 expression level was controlled by several mechanisms. However, the regulation mechanism of cardiac PHLPP1 expression following myocardial I/R remains unknown. MAIN METHODS The current study utilized the mouse model of myocardial I/R injury in vivo and the neonatal rat ventricular myocytes (NRVMs) of hypoxia/reoxygenation (H/R) injury in vitro. Expression of PHLPP1, nuclear factor-kappa B (NF-κB) and pNF-κB were determined by western blot. The expression of PHLPP1 and translocation of NF-κB was assessed by immunofluorescence. Chromatin immunoprecipitation (ChIP) assay was used to detect the binding of NF-κB to the promoter region of phlpp1 gene. KEY FINDINGS Myocardial I/R had no effect on cardiac PHLPP1 expression following I/R (30 min/2 h) but decreased after 4 h reperfusion. In vitro, H/R (4 h/1 h) and tumor necrosis factor-alpha (TNF-α)-stimulation resulted in upregulation of PHLPP1 in NRVMs, which was blocked with etanercept. Yet, H2O2-induced oxidative stress had no obvious effect on PHLPP1 expression of NRVMs at early stage but N-acetylcysteine (NAC) pretreatment increased PHLPP1 levels after 4 h H2O2 stimulation. TNF-α and H/R led to both expression and transcriptional activity of NF-κB, accompany with higher expression of PHLPP1. Pyrrolidine dithiocarbamate (PDTC), a NF-κB inhibitor, prevented the response not only in TNF-α-treated cardiomyocytes but also in H/R-treated group. SIGNIFICANCE These results implicated that TNF-α involved in cardiac PHLPP1 upregulation during reoxygenation, which was mediated by NF-κB transcriptional activity.
Collapse
Affiliation(s)
- Bin Tang
- Department of International Medical, China-Japan Friendship Hospital, Beijing 100029, China
| | - Jing Ma
- Department of Traditional Chinese Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Xiaoqin Ha
- Department of Clinical Laboratory Medicine, Lanzhou General Hospital of Lanzhou Military Region, People's Liberation Army, Key Laboratory of Stem Cell and Gene Drug in Gansu Province, Lanzhou 730000, China
| | - Yuanqiang Zhang
- Department of Histology and Embryology, Fourth Military Medical University, Xi'an 710032, China.
| | - Yuan Xing
- Department of Clinical Laboratory Medicine, Lanzhou General Hospital of Lanzhou Military Region, People's Liberation Army, Key Laboratory of Stem Cell and Gene Drug in Gansu Province, Lanzhou 730000, China.
| |
Collapse
|
20
|
Alpha7 nicotinic acetylcholine receptor activation protects against myocardial reperfusion injury through modulation of autophagy. Biochem Biophys Res Commun 2018; 500:357-364. [DOI: 10.1016/j.bbrc.2018.04.077] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 04/10/2018] [Indexed: 12/29/2022]
|
21
|
Mathur A, Pandey VK, Kakkar P. Activation of GSK3β/β-TrCP axis via PHLPP1 exacerbates Nrf2 degradation leading to impairment in cell survival pathway during diabetic nephropathy. Free Radic Biol Med 2018; 120:414-424. [PMID: 29655866 DOI: 10.1016/j.freeradbiomed.2018.04.550] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 03/27/2018] [Accepted: 04/11/2018] [Indexed: 01/06/2023]
Abstract
NF-E2 p45-related factor 2 (Nrf2), is a major redox sensitive transcription factor that plays an essential role in regulating glucose metabolism. Inactivation of Nrf2 has been associated with diabetic complications however, mechanisms warranting Nrf2 suppression are incompletely understood. We hypothesized that PHLPP1 activates GSK3β to induce β-TrCP mediated Nrf2 phosphorylation and degradation. In vivo study was carried out in STZ-NA induced type 2 diabetic male Wistar rats. GSK3β mediated Nrf2 ubiquitination was confirmed by administration of GSK3β inhibitor (LiCl; 60 mg/kg bwt.) which rapidly enhanced Nrf2 protein levels in STZ-NA treated diabetic rats. In addition, high glucose (30 mM; 48 h) treated renal proximal tubular cells NRK52E showed decreased Nrf2 nuclear localization, enhanced oxidative stress and caspase3 activation. While specific inhibition with GSK3β inhibitor SB216763 in vitro restored cellular homeostasis, glucose uptake and decreased apoptotic cell death. Immunoblotting and immunocytochemistry data demonstrated that aberrant renal glucose fluxes are associated with p53 mediated modulation in glucose transporter levels where expression of p53 is indirectly targeted through Nrf2 responsive MDM2 protein. Gene knockdown of PHLPP1 in NRK52E cells enhanced Nrf2-responsive antioxidant enzymes HO-1 and NQO-1 which suggested that PHLPP1 up-regulation during hyperglycemia lowers Nrf2 stability via GSK3β activation. More significantly, GSK3β inhibition enhanced Nrf2-ARE binding compared to diabetic rats, providing further confirmation for GSK3β/β-TrCP pathway in suppressing Nrf2 activation during diabetic renal injury. Taken together, our results indicate that PHLPP1 up-surged Nrf2 nuclear instability by promoting Nrf2/β-TrCP association and its inhibition may be critical in the management of diabetic nephropathy.
Collapse
Affiliation(s)
- Alpana Mathur
- Herbal Research Laboratory, Food, Drug & Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; Babu Banarasi Das University, Lucknow, Uttar Pradesh, India
| | - Vivek Kumar Pandey
- Herbal Research Laboratory, Food, Drug & Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research, CSIR-IITR Campus, Lucknow, Uttar Pradesh, India
| | - Poonam Kakkar
- Herbal Research Laboratory, Food, Drug & Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; Babu Banarasi Das University, Lucknow, Uttar Pradesh, India; Academy of Scientific and Innovative Research, CSIR-IITR Campus, Lucknow, Uttar Pradesh, India.
| |
Collapse
|
22
|
Hwang SM, Feigenson M, Begun DL, Shull LC, Culley KL, Otero M, Goldring MB, Ta LE, Kakar S, Bradley EW, Westendorf JJ. Phlpp inhibitors block pain and cartilage degradation associated with osteoarthritis. J Orthop Res 2018; 36:1487-1497. [PMID: 29068480 PMCID: PMC5985222 DOI: 10.1002/jor.23781] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 10/21/2017] [Indexed: 02/04/2023]
Abstract
Phlpp protein phosphatases are abnormally abundant within human osteoarthritic articular chondrocytes and may contribute to the development of osteoarthritis. Mice lacking Phlpp1 were previously shown to be resistant to post-traumatic osteoarthritis. Here a small molecule with therapeutic properties that inhibits Phlpp1 and Phlpp2 was tested for its ability to slow post-traumatic OA in mice and to stimulate anabolic pathways in human articular cartilage from OA joints. PTOA was induced in male C57Bl/6 mice by surgically destabilizing the meniscus. Seven weeks after surgery, mice received a single intra-articular injection of the Phlpp inhibitor NSC117079 or saline. Mechanical allodynia was measured with von Frey assays, mobility was tracked in an open field system, and cartilage damage was assessed histologically. A single intra-articular injection of the Phlpp inhibitor NSC117079 attenuated mechanical allodynia and slowed articular cartilage degradation in joints with a destabilized meniscus. Animals treated with the Phlpp inhibitor 7 weeks after injury maintained normal activity levels, while those in the control group traveled shorter distances and were less active 3 months after the joint injury. NSC117079 also increased production of cartilage extracellular matrix components (glycosaminoglycans and aggrecan) in over 90% of human articular cartilage explants from OA patients and increased phosphorylation of Phlpp1 substrates (AKT2, ERK1/2, and PKC) in human articular chondrocytes. Our results indicate that Phlpp inhibitor NSC117079 is a novel osteoarthritis disease modifying drug candidate that may have palliative affects. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:1487-1497, 2018.
Collapse
Affiliation(s)
- Soyun M. Hwang
- Mayo Clinic School of Medicine, Mayo Clinic, Rochester, MN, 55905 USA
| | - Marina Feigenson
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905 USA
| | - Dana L. Begun
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905 USA
| | | | - Kirsty L. Culley
- HSS Research Institute, Hospital for Special Surgery, Weill Cornell Medical College, New York, NY 10021, USA
| | - Miguel Otero
- HSS Research Institute, Hospital for Special Surgery, Weill Cornell Medical College, New York, NY 10021, USA
| | - Mary B. Goldring
- HSS Research Institute, Hospital for Special Surgery, Weill Cornell Medical College, New York, NY 10021, USA
| | - Lauren E. Ta
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905 USA
| | - Sanjeev Kakar
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905 USA
| | - Elizabeth W. Bradley
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905 USA,Co-corresponding authors. ,
| | - Jennifer J. Westendorf
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905 USA,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905 USA,Co-corresponding authors. ,
| |
Collapse
|
23
|
Qiu Y, Wu Y, Meng M, Luo M, Zhao H, Sun H, Gao S. GYY4137 protects against myocardial ischemia/reperfusion injury via activation of the PHLPP-1/Akt/Nrf2 signaling pathway in diabetic mice. J Surg Res 2018; 225:29-39. [PMID: 29605032 DOI: 10.1016/j.jss.2017.12.030] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 11/21/2017] [Accepted: 12/19/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND This study explores the protective effects of a hydrogen sulfide donor, morpholin-4-ium 4-methoxyphenyl-morpholino-phosphinodithioate (GYY4137), in the hearts of diabetic mice that had been subjected to myocardial ischemia/reperfusion injury. Diabetes impairs the Akt pathway, in which the Akt protein is dephosphorylated and inactivated by PH domain leucine-rich repeat protein phosphatase-1 (PHLPP-1). However, the function of PHLPP-1 and molecular mechanism that underlies the cardiac protection exerted by GYY4137 remains unknown. METHODS Diabetic or nondiabetic mice were subjected to 45 min of coronary artery occlusion followed by 2 h of reperfusion. H9c2 cells were cultured with normal or high glucose and then subjected to 3 h of hypoxia followed by 6 h of reoxygenation. Pretreatment with GYY4137 was performed in a randomized manner before ischemia/reperfusion or hypoxia/reoxygenation. The infarct size, cardiomyocyte apoptosis, and oxidative stress were measured. Western blotting was conducted to elucidate the protective mechanism. RESULTS Diabetic mice or H9c2 cells exposed to high glucose displayed a larger infarct size, more severe cardiomyocyte apoptosis, lower cell viability, and increased oxidative stress, which were associated with increased levels of PHLPP-1 and reduced levels of p-Akt and nuclear factor-erythroid-2-related factor 2 (Nrf2) protein expression. These changes were prevented/reversed by GYYG4137 pretreatment. At the cellular level, PHLPP-1 siRNA attenuated cellular injury, and this was associated with increased p-Akt and nuclear Nrf2 protein, whereas the decrement of Akt phosphorylation induced by LY294002 augmented cellular injury and decreased nuclear Nrf2. CONCLUSIONS GYY4137 activates the PHLPP-1/Akt/Nrf2 pathway to protect against diabetic myocardial ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Yun Qiu
- Department of Emergency Medicine, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu Province, China
| | - Yichen Wu
- Department of Emergency Medicine, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu Province, China
| | - Min Meng
- Department of Emergency Medicine, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu Province, China
| | - Man Luo
- Department of Emergency Medicine, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu Province, China
| | - Hongmei Zhao
- Department of Emergency Medicine, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu Province, China
| | - Hong Sun
- Department of Emergency Medicine, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu Province, China
| | - Sumin Gao
- Department of Emergency Medicine, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu Province, China.
| |
Collapse
|
24
|
Guo J, Zhang S, Ma L, Shi H, Zhu J, Wu J, An Y, Ge J. Cardioprotection by Mild Hypothermia Is Abolished in Aged Mice. Ther Hypothermia Temp Manag 2017; 7:193-198. [PMID: 28445087 DOI: 10.1089/ther.2017.0001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Junjie Guo
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao University, Shandong, China
| | - Shuning Zhang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Leilei Ma
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hongtao Shi
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jianbing Zhu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Wu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yi An
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao University, Shandong, China
| | - Junbo Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
25
|
Li R, Fang J, Huo B, Su YS, Wang J, Liu LG, Hu M, Cheng C, Zheng P, Zhu XH, Jiang DS, Wei X. Leucine-rich repeat neuronal protein 4 (LRRN4) potentially functions in dilated cardiomyopathy. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2017; 10:9925-9933. [PMID: 31966882 PMCID: PMC6965904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 08/23/2017] [Indexed: 06/10/2023]
Abstract
Leucine-rich repeat neuronal protein-4 (LRRN4 or NLRR4) has been identified as a new member of LRRN family, which is a group of proteins that contain leucine-rich repeat domains and functioned as regulators in a variety of pathologic processes including cardiac remodeling. However, the exact pattern of expression and function of LRRN4 in the human hearts is still unclear. In our study, the western blot test and real-time PCR were performed to detect the LRRN4 level in hearts of patients with dilated cardiomyopathy (DCM), ischemia heart disease (IHD) hearts respectively. Interestingly, the LRRN4 was highly expressed in donor hearts, but significantly reduced in hearts with DCM. While a comparable level of expression was detected in the IHD hearts when compared with donor hearts. Immunohistochemistry assay showed that LRRN4 was particularly expressed in cardiomyocytes and responsible for its decreased expression in the DCM hearts. Furthermore, we found LRRN4 was expressed in the ventricular cardiomyocytes of mice and apparently reduced after pressure overload treatment in the wild type mice. Therefore, our hitherto unrecognized findings provided the first evidence that the highly expressed LRRN4 is critical for maintaining morphology and function of heart. In addition to that, since its expression level decreased in DCM hearts but not IHD hearts, which indicated LRRN4 might be a therapeutic target clinically for DCM disease.
Collapse
Affiliation(s)
- Rui Li
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
- Heart-Lung Transplantation Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
- Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
| | - Jing Fang
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
- Heart-Lung Transplantation Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
- Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
| | - Bo Huo
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
- Heart-Lung Transplantation Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
- Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
| | - Yun-Shu Su
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
- Heart-Lung Transplantation Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
- Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
| | - Jing Wang
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
- Heart-Lung Transplantation Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
- Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
| | - Li-Gang Liu
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
| | - Min Hu
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
| | - Cai Cheng
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
| | - Ping Zheng
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
| | - Xue-Hai Zhu
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
- Key Laboratory of Organ Transplantation, Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
| | - Ding-Sheng Jiang
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
- Key Laboratory of Organ Transplantation, Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
| | - Xiang Wei
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
- Key Laboratory of Organ Transplantation, Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
| |
Collapse
|
26
|
Mathur A, Pandey VK, Kakkar P. PHLPP: a putative cellular target during insulin resistance and type 2 diabetes. J Endocrinol 2017; 233:R185-R198. [PMID: 28428363 DOI: 10.1530/joe-17-0081] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 04/20/2017] [Indexed: 12/29/2022]
Abstract
Progressive research in the past decade converges to the impact of PHLPP in regulating the cellular metabolism through PI3K/AKT inhibition. Aberrations in PKB/AKT signaling coordinates with impaired insulin secretion and insulin resistance, identified during T2D, obesity and cardiovascular disorders which brings in the relevance of PHLPPs in the metabolic paradigm. In this review, we discuss the impact of PHLPP isoforms in insulin signaling and its associated cellular events including mitochondrial dysfunction, DNA damage, autophagy and cell death. The article highlights the plausible molecular targets that share the role during insulin-resistant states, whose understanding can be extended into treatment responses to facilitate targeted drug discovery for T2D and allied metabolic syndromes.
Collapse
Affiliation(s)
- Alpana Mathur
- Herbal Research LaboratoryCSIR-Indian Institute of Toxicology Research, Lucknow, India
- Babu Banarasi Das UniversityBBD City, Lucknow, India
| | - Vivek Kumar Pandey
- Herbal Research LaboratoryCSIR-Indian Institute of Toxicology Research, Lucknow, India
- Academy of Scientific and Innovative ResearchCSIR-IITR, Lucknow, India
| | - Poonam Kakkar
- Herbal Research LaboratoryCSIR-Indian Institute of Toxicology Research, Lucknow, India
- Babu Banarasi Das UniversityBBD City, Lucknow, India
- Academy of Scientific and Innovative ResearchCSIR-IITR, Lucknow, India
| |
Collapse
|
27
|
Nuclear AMPK regulated CARM1 stabilization impacts autophagy in aged heart. Biochem Biophys Res Commun 2017; 486:398-405. [PMID: 28315332 DOI: 10.1016/j.bbrc.2017.03.053] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 03/13/2017] [Indexed: 12/27/2022]
Abstract
Senescence-associated autophagy downregulation leads to cardiomyocyte dysfunction. Coactivator-associated arginine methyltransferase 1 (CARM1) participates in many cellular processes, including autophagy in mammals. However, the effect of CARM1 in aging-related cardiac autophagy decline remains undefined. Moreover, AMP-activated protein kinase (AMPK) is a key regulator in metabolism and autophagy, however, the role of nuclear AMPK in autophagy outcome in aged hearts still unclear. Hers we identify the correlation between nuclear AMPK and CARM1 in aging heart. We found that fasting could promote autophagy in young hearts but not in aged hearts. The CARM1 stabilization is markedly decrease in aged hearts, which impaired nucleus TFEB-CARM1 complex and autophagy flux. Further, S-phase kinase-associated protein 2(SKP2), responsible for CARM1 degradation, was increased in aged hearts. We further validated that AMPK dependent FoxO3 phosphorylation was markedly reduced in nucleus, the decreased nuclear AMPK-FoxO3 activity fails to suppress SKP2-E3 ubiquitin ligase. This loss of repression leads to The CARM1 level and autophagy in aged hearts could be restored through AMPK activation. Taken together, AMPK deficiency results in nuclear CARM1 decrease mediated in part by SKP2, contributing to autophagy dysfunction in aged hearts. Our results identified nuclear AMPK controlled CARM1 stabilization as a new actor that regulates cardiac autophagy.
Collapse
|
28
|
Signaling Pathways in Cardiac Myocyte Apoptosis. BIOMED RESEARCH INTERNATIONAL 2016; 2016:9583268. [PMID: 28101515 PMCID: PMC5215135 DOI: 10.1155/2016/9583268] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 11/20/2016] [Indexed: 12/16/2022]
Abstract
Cardiovascular diseases, the number 1 cause of death worldwide, are frequently associated with apoptotic death of cardiac myocytes. Since cardiomyocyte apoptosis is a highly regulated process, pharmacological intervention of apoptosis pathways may represent a promising therapeutic strategy for a number of cardiovascular diseases and disorders including myocardial infarction, ischemia/reperfusion injury, chemotherapy cardiotoxicity, and end-stage heart failure. Despite rapid growth of our knowledge in apoptosis signaling pathways, a clinically applicable treatment targeting this cellular process is currently unavailable. To help identify potential innovative directions for future research, it is necessary to have a full understanding of the apoptotic pathways currently known to be functional in cardiac myocytes. Here, we summarize recent progress in the regulation of cardiomyocyte apoptosis by multiple signaling molecules and pathways, with a focus on the involvement of these pathways in the pathogenesis of heart disease. In addition, we provide an update regarding bench to bedside translation of this knowledge and discuss unanswered questions that need further investigation.
Collapse
|