1
|
Saloman JL, Epouhe AY, Ruff CF, Albers KM. PDX1, a transcription factor essential for organ differentiation, regulates SERCA-dependent Ca 2+ homeostasis in sensory neurons. Cell Calcium 2024; 120:102884. [PMID: 38574509 PMCID: PMC11188734 DOI: 10.1016/j.ceca.2024.102884] [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: 01/05/2024] [Revised: 03/13/2024] [Accepted: 03/31/2024] [Indexed: 04/06/2024]
Abstract
Pancreatic and duodenal homeobox 1 (PDX1) is a transcription factor required for the development and differentiation of the pancreas. Previous studies indicated that PDX1 expression was restricted to the gastrointestinal tract. Using a cre-dependent reporter, we observed PDX1-dependent expression of tdtomato (PDX1-tom) in a subpopulation of sensory nerves. Many of these PDX1-tom afferents expressed the neurofilament 200 protein and projected to the skin. Tdtomato-labeled terminals were associated with hair follicles in the form of longitudinal and circumferential lanceolate endings suggesting a role in tactile and proprioceptive perception. To begin to examine the functional significance of PDX1 in afferents, we used Fura-2 imaging to examine calcium (Ca2+) handling under naïve and nerve injury conditions. Neuropathic injury is associated with increased intracellular Ca2+ signaling that in part results from dysregulation of the sarco/endoplasmic reticulum calcium transport ATPase (SERCA). Here we demonstrate that under naïve conditions, PDX1 regulates expression of the SERCA2B isoform in sensory neurons. In response to infraorbital nerve injury, a significant reduction of PDX1 and SERCA2B expression and dysregulation of Ca2+ handling occurs in PDX1-tom trigeminal ganglia neurons. The identification of PDX1 expression in the somatosensory system and its regulation of SERCA2B and Ca2+ handling provide a new mechanism to explain pathological changes in primary afferents that may contribute to pain associated with nerve injury.
Collapse
Affiliation(s)
- Jami L Saloman
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Neurobiology, Center for Neuroscience and Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Ariel Y Epouhe
- Department of Neurobiology, Center for Neuroscience and Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Catherine F Ruff
- Department of Neurobiology, Center for Neuroscience and Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kathryn M Albers
- Department of Neurobiology, Center for Neuroscience and Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
2
|
Romero-Becerra R, Cruz FM, Mora A, Lopez JA, Ponce-Balbuena D, Allan A, Ramos-Mondragón R, González-Terán B, León M, Rodríguez ME, Leiva-Vega L, Guerrero-Serna G, Jimenez-Vazquez EN, Filgueiras-Rama D, Vázquez J, Jalife J, Sabio G. p38γ/δ activation alters cardiac electrical activity and predisposes to ventricular arrhythmia. NATURE CARDIOVASCULAR RESEARCH 2023; 2:1204-1220. [PMID: 39196141 DOI: 10.1038/s44161-023-00368-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 10/19/2023] [Indexed: 08/29/2024]
Abstract
Ventricular fibrillation (VF) is a leading immediate cause of sudden cardiac death. There is a strong association between aging and VF, although the mechanisms are unclear, limiting the availability of targeted therapeutic interventions. Here we found that the stress kinases p38γ and p38δ are activated in the ventricles of old mice and mice with genetic or drug-induced arrhythmogenic conditions. We discovered that, upon activation, p38γ and p38δ cooperatively increase the susceptibility to stress-induced VF. Mechanistically, our data indicate that activated p38γ and p38δ phosphorylate ryanodine receptor 2 (RyR2) disrupt Kv4.3 channel localization, promoting sarcoplasmic reticulum calcium leak, Ito current reduction and action potential duration prolongation. In turn, this led to aberrant intracellular calcium handling, premature ventricular complexes and enhanced susceptibility to VF. Blocking this pathway protected genetically modified animals from VF development and reduced the VF duration in aged animals. These results indicate that p38γ and p38δ are a potential therapeutic target for sustained VF prevention.
Collapse
Affiliation(s)
| | - Francisco M Cruz
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Alfonso Mora
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Juan Antonio Lopez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Daniela Ponce-Balbuena
- Center for Arrhythmia Research, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Andrew Allan
- Center for Arrhythmia Research, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Roberto Ramos-Mondragón
- Center for Arrhythmia Research, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Bárbara González-Terán
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Gladstone Institutes, San Francisco, CA, USA
| | - Marta León
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | | - Luis Leiva-Vega
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Guadalupe Guerrero-Serna
- Center for Arrhythmia Research, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Eric N Jimenez-Vazquez
- Center for Arrhythmia Research, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - David Filgueiras-Rama
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Hospital Clínico Universitario San Carlos, Madrid, Spain
| | - Jesús Vázquez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - José Jalife
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.
- Center for Arrhythmia Research, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.
| | - Guadalupe Sabio
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| |
Collapse
|
3
|
New insights into the mechanism of freeze-induced damage based on ice crystal morphology and exudate proteomics. Food Res Int 2022; 161:111757. [DOI: 10.1016/j.foodres.2022.111757] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/21/2022] [Accepted: 07/27/2022] [Indexed: 11/17/2022]
|
4
|
Cholecystokinin Octapeptide Promotes ANP Secretion through Activation of NOX4-PGC-1 α-PPAR α/PPAR γ Signaling in Isolated Beating Rat Atria. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5905374. [PMID: 35770043 PMCID: PMC9236793 DOI: 10.1155/2022/5905374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 05/15/2022] [Accepted: 05/19/2022] [Indexed: 11/18/2022]
Abstract
Atrial natriuretic peptide (ANP), a canonical cardiac hormone, is mainly secreted from atrial myocytes and is involved in the regulation of body fluid, blood pressure homeostasis, and antioxidants. Cholecystokinin (CCK) is also found in cardiomyocytes as a novel cardiac hormone and induces multiple cardiovascular regulations. However, the direct role of CCK on the atrial mechanical dynamics and ANP secretion is unclear. The current study was to investigate the effect of CCK octapeptide (CCK-8) on the regulation of atrial dynamics and ANP secretion. Experiments were performed in isolated perfused beating rat atria. ANP was measured using radioimmunoassay. The levels of hydrogen peroxide (H2O2) and arachidonic acid (AA) were determined using ELISA Kits. The levels of relative proteins and mRNA were detected by Western blot and RT-qPCR. The results showed that sulfated CCK-8 (CCK-8s) rather than desulfated CCK-8 increased the levels of phosphorylated cytosolic phospholipase A2 and AA release through activation of CCK receptors. This led to the upregulation of NADPH oxidase 4 (NOX4) expression levels and H2O2 production and played a negative inotropic effect on atrial mechanical dynamics via activation of ATP-sensitive potassium channels and large-conductance calcium-activated potassium channels. In addition, CCK-8s-induced NOX4 subsequently upregulated peroxisome proliferator-activated receptor γ (PPARγ) coactivator-1α (PGC-1α) expression levels through activation of p38 mitogen-activated protein kinase as well as the serine/threonine kinase signaling, ultimately promoting the secretion of ANP via activation of PPARα and PPARγ. In the presence of the ANP receptor inhibitor, the CCK-8-induced increase of AA release, H2O2 production, and the upregulation of NOX4 and CAT expressions was augmented but the SOD expression induced by CCK-8s was repealed. These findings indicate that CCK-8s promotes the secretion of ANP through activation of NOX4-PGC-1α-PPARα/PPARγ signaling, in which ANP is involved in resistance for NOX4 expression and ROS production and regulation of SOD expression.
Collapse
|
5
|
Yang Q, Chan P. Skeletal Muscle Metabolic Alternation Develops Sarcopenia. Aging Dis 2022; 13:801-814. [PMID: 35656108 PMCID: PMC9116905 DOI: 10.14336/ad.2021.1107] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 11/28/2021] [Indexed: 11/23/2022] Open
Abstract
Sarcopenia is a new type of senile syndrome with progressive skeletal muscle mass loss with age, accompanied by decreased muscle strength and/or muscle function. Sarcopenia poses a serious threat to the health of the elderly and increases the burden of family and society. The underlying pathophysiological mechanisms of sarcopenia are still unclear. Recent studies have shown that changes of skeletal muscle metabolism are the risk factors for sarcopenia. Furthermore, the importance of the skeletal muscle metabolic microenvironment in regulating satellite cells (SCs) is gaining significant attention. Skeletal muscle metabolism has intrinsic relationship with the regulation of skeletal muscle mass and regeneration. This review is to discuss recent findings regarding skeletal muscle metabolic alternation and the development of sarcopenia, hoping to contribute better understanding and treatment of sarcopenia.
Collapse
Affiliation(s)
- Qiumei Yang
- Department of Neurology, Geriatrics and Neurobiology, National Clinical Research Center of Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing, China.
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China.
| | - Piu Chan
- Department of Neurology, Geriatrics and Neurobiology, National Clinical Research Center of Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing, China.
- Clinical Center for Parkinson’s Disease, Capital Medical University, Beijing Institute of Geriatrics, Beijing, China.
- Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory for Parkinson’s Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China.
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China.
- Correspondence should be addressed to: Dr. Piu Chan, Department of Neurobiology, Xuanwu Hospital of Capital Medical University, 45 Changchun Road, Beijing 100053, China. .
| |
Collapse
|
6
|
Changes in Thyroid Hormone Signaling Mediate Cardiac Dysfunction in the Tg197 Mouse Model of Arthritis: Potential Therapeutic Implications. J Clin Med 2021; 10:jcm10235512. [PMID: 34884213 PMCID: PMC8658216 DOI: 10.3390/jcm10235512] [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: 11/01/2021] [Revised: 11/21/2021] [Accepted: 11/24/2021] [Indexed: 11/17/2022] Open
Abstract
Background Rheumatoid Arthritis (RA) patients show a higher risk of heart failure. The present study investigated possible causes of cardiac dysfunction related to thyroid hormone (TH) signaling in a RA mouse model. Methods A TNF-driven mouse model of RA[TghuTNF (Tg197)] was used. Cardiac function was evaluated by echocardiography. SERCA2a and phospholamban protein levels in left ventricle (LV) tissue, thyroid hormone levels in serum, TH receptors in LV and TH-related kinase signaling pathways were measured. T3 hormone was administered in female Tg197 mice. Results We show LV and atrial dilatation with systolic dysfunction in Tg197 animals, accompanied by downregulated SERCA2a. We suggest an interaction of pro-inflammatory and thyroid hormone signaling indicated by increased p38 MAPK and downregulation of TRβ1 receptor in Tg197 hearts. Interestingly, female Tg197 mice showed a worse cardiac phenotype related to reduced T3 levels and Akt activation. T3 supplementation increased Akt activation, restored SERCA2a expression and improved cardiac function in female Tg197 mice. Conclusions TNF overexpression of Tg197 mice results in cardiac dysfunction via p38 MAPK activation and downregulation of TRβ1. Gender-specific reduction in T3 levels could cause the worse cardiac phenotype observed in female mice, while T3 administration improves cardiac function and calcium handling via modified Akt activation.
Collapse
|
7
|
Jing J, Jiang X, Zhu C, Zheng Q, Ji Q, Yin H, Huang J, Zhu Y, Wang J, Qin S, Ling Y. Dynamic changes of miRNAs in skeletal muscle development at New Zealand rabbits. BMC Genomics 2021; 22:577. [PMID: 34315409 PMCID: PMC8314457 DOI: 10.1186/s12864-021-07896-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 07/08/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND miRNA is one of the crucial roles in the complex and dynamic network that regulates the development of skeletal muscle. The landscape of skeletal muscle miRNAs from fetus to adult in New Zealand rabbits has not been revealed yet. RESULTS In this study, nine RNA-seq libraries of fetus, child and adult rabbits' leg muscles were constructed. A total of 278 differentially expressed miRNAs (DEmiRNAs) were identified. In the fetus vs. child group, the main functional enrichments were involved in membrane and transport. Pathway enriched terms of up-regulated DEmiRNAs were connected with the differentiation and hypertrophy of skeletal muscle, and down-regulated ones were related to muscle structure and metabolic capacity. In the child vs. adult group, functions were associated to positioning and transportation, and pathways were relevant to ECM, muscle structure and hypertrophy. Finally, ocu-miR-185-3p and ocu-miR-370-3p, which had the most target genes, were identified as hub-miRNAs in these two groups. CONCLUSIONS In short, we summarized the highly expressed and uniquely expressed DEmiRNAs of fetus, child and adult rabbits' leg muscles. Besides, the potential functional changes of miRNAs in two consecutive stages have been explored. Among them, the ocu-miR-185-3p and ocu-miR-370-3p with the most target genes were selected as hub-miRNAs. These data improved the understanding of the regulatory molecules of meat rabbit development, and provided a novel perspective for molecular breeding of meat rabbits.
Collapse
Affiliation(s)
- Jing Jing
- College of Animal Science and Technology, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China.,Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio- Breeding, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China
| | - Xichun Jiang
- Institute of Animal Science and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Anhui, 230031, Hefei, People's Republic of China
| | - Cuiyun Zhu
- College of Animal Science and Technology, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China.,Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio- Breeding, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China
| | - Qi Zheng
- College of Animal Science and Technology, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China.,Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio- Breeding, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China
| | - Qianyun Ji
- College of Animal Science and Technology, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China.,Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio- Breeding, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China
| | - Huiqun Yin
- Reproductive Medicine Center, The 901st Hospital, Anhui, 230031, Hefei, People's Republic of China
| | - Jingtong Huang
- College of Animal Science and Technology, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China.,Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio- Breeding, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China
| | - Yixiao Zhu
- College of Animal Science and Technology, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China.,Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio- Breeding, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China
| | - Jiao Wang
- College of Animal Science and Technology, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China.,Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio- Breeding, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China
| | - Shuaiqi Qin
- College of Animal Science and Technology, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China.,Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio- Breeding, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China
| | - Yinghui Ling
- College of Animal Science and Technology, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China. .,Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio- Breeding, Anhui Agricultural University, Anhui, 230036, Hefei, People's Republic of China.
| |
Collapse
|
8
|
Men XM, Xu ZW, Tao X, Deng B, Qi KK. FNDC5 expression closely correlates with muscle fiber types in porcine longissimus dorsi muscle and regulates myosin heavy chains (MyHCs) mRNA expression in C2C12 cells. PeerJ 2021; 9:e11065. [PMID: 33976958 PMCID: PMC8061570 DOI: 10.7717/peerj.11065] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 02/15/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Irisin (a glycosylated protein) is cleaved from fibronectin type III domain-containing protein 5 (FNDC5), which is expressed mainly in animal muscle tissues and has multiple metabolic regulatory activities. However, their roles in controlling myofiber types in skeletal muscle remain unclear. METHODOLOGY Two different commercial hybridized pigs, LJH (a crossed pig containing Chinese native pig genotypes) and DLY (Duroc × Landrace × Yorkshire) were selected to analyze FNDC5 mRNA expression and the mRNA composition of four adult myosin heavy chain (MyHC) isoforms (IIIaIIxIIb) in the longissimus dorsi (LD) muscle. C2C12 myoblasts were cultured to investigate the effects of FNDC5 on the four MyHCs mRNA expressive levels, using small interfering RNA for depletion and a eukaryotic expression vector carrying FNDC5 for overexpression. ZLN005 (a small molecule activator of FNDC5's upstream control gene PGC1α) or recombinant human irisin protein were also used. RESULTS In LD muscle, LJH pigs had the higher FNDC5 mRNA level, and MyHC I or IIa proportion than DLY pigs (P < 0.05). For C2C12 cells in vitro, small interfering RNA (si-592) silencing of FNDC5 expression markedly reduced MyHC IIa mRNA levels (P < 0.05), while FNDC5 overexpression significantly increased MyHC IIa mRNA levels (P < 0.05). Exogenous irisin increased the mRNA levels of PGC1α (peroxisome proliferator-activated receptor gamma coactivator 1-alpha), FNDC5, MyHCI, MyHCIIa, NRF1 (nuclear respiratory factor 1), VEGF (vascular endothelial growth factor), and TFAM (mitochondrial transcription factor A,) (P < 0.05), and the enzyme activities of SDH (succinate dehydrogenase), CK (creatine kinase), and MDH (malate dehydrogenase) in C2C12 myotubes (P < 0.05). These results showed that FNDC5 mRNA expression had a significant association with the characteristics of myofiber types in porcine muscle, and participated in regulating MyHCs mRNA expression of C2C12 myogenic differentiation cells in vitro. FNDC5 could be an important factor to control muscle fiber types, which provides a new direction to investigate pork quality via muscle fiber characteristics.
Collapse
Affiliation(s)
- Xiao-Ming Men
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Science, Hangzhou, Zhejiang, China
| | - Zi-Wei Xu
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Science, Hangzhou, Zhejiang, China
| | - Xin Tao
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Science, Hangzhou, Zhejiang, China
| | - Bo Deng
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Science, Hangzhou, Zhejiang, China
| | - Ke-Ke Qi
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Science, Hangzhou, Zhejiang, China
| |
Collapse
|
9
|
Mekies LN, Regev D, Eisen B, Fernandez‐Gracia J, Baskin P, Ben Jehuda R, Shulman R, Reiter I, Palty R, Arad M, Gottlieb E, Binah O. Depressed β-adrenergic inotropic responsiveness and intracellular calcium handling abnormalities in Duchenne Muscular Dystrophy patients' induced pluripotent stem cell-derived cardiomyocytes. J Cell Mol Med 2021; 25:3922-3934. [PMID: 33619882 PMCID: PMC8051742 DOI: 10.1111/jcmm.16341] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/05/2021] [Accepted: 01/14/2021] [Indexed: 12/20/2022] Open
Abstract
Duchenne muscular dystrophy (DMD), caused by mutations in the dystrophin gene, is an X-linked disease affecting male and rarely adult heterozygous females, resulting in death by the late 20s to early 30s. Previous studies reported depressed left ventricular function in DMD patients which may result from deranged intracellular Ca2+ -handling. To decipher the mechanism(s) underlying the depressed LV function, we tested the hypothesis that iPSC-CMs generated from DMD patients feature blunted positive inotropic response to β-adrenergic stimulation. To test the hypothesis, [Ca2+ ]i transients and contractions were recorded from healthy and DMD-CMs. While in healthy CMs (HC) isoproterenol caused a prominent positive inotropic effect, DMD-CMs displayed a blunted inotropic response. Next, we tested the functionality of the sarcoplasmic reticulum (SR) by measuring caffeine-induced Ca2+ release. In contrast to HC, DMD-CMs exhibited reduced caffeine-induced Ca2+ signal amplitude and recovery time. In support of the depleted SR Ca2+ stores hypothesis, in DMD-CMs the negative inotropic effects of ryanodine and cyclopiazonic acid were smaller than in HC. RNA-seq analyses demonstrated that in DMD CMs the RNA-expression levels of specific subunits of the L-type calcium channel, the β1-adrenergic receptor (ADRβ1) and adenylate cyclase were down-regulated by 3.5-, 2.8- and 3-fold, respectively, which collectively contribute to the depressed β-adrenergic responsiveness.
Collapse
MESH Headings
- Adrenergic Agents/pharmacology
- Adult
- Calcium/metabolism
- Calcium Channels, L-Type/genetics
- Calcium Channels, L-Type/metabolism
- Cell Differentiation
- Female
- Gene Expression Regulation
- Humans
- Induced Pluripotent Stem Cells/drug effects
- Induced Pluripotent Stem Cells/metabolism
- Induced Pluripotent Stem Cells/pathology
- Male
- Middle Aged
- Muscular Dystrophy, Duchenne/drug therapy
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Myocardial Contraction
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- RNA-Seq
- Receptors, Adrenergic, beta-1/genetics
- Receptors, Adrenergic, beta-1/metabolism
- Sarcoplasmic Reticulum/drug effects
- Sarcoplasmic Reticulum/metabolism
- Sarcoplasmic Reticulum/pathology
Collapse
Affiliation(s)
- Lucy N. Mekies
- Department of PhysiologyBiophysics and Systems BiologyRappaport Faculty of MedicineTechnion – Israel Institute of TechnologyHaifaIsrael
| | - Danielle Regev
- Department of PhysiologyBiophysics and Systems BiologyRappaport Faculty of MedicineTechnion – Israel Institute of TechnologyHaifaIsrael
| | - Binyamin Eisen
- Department of PhysiologyBiophysics and Systems BiologyRappaport Faculty of MedicineTechnion – Israel Institute of TechnologyHaifaIsrael
| | - Jonatan Fernandez‐Gracia
- Department of Cell Biology and Cancer ScienceRappaport Faculty of MedicineTechnion – Israel Institute of TechnologyHaifaIsrael
| | - Polina Baskin
- Department of PhysiologyBiophysics and Systems BiologyRappaport Faculty of MedicineTechnion – Israel Institute of TechnologyHaifaIsrael
| | - Ronen Ben Jehuda
- Department of PhysiologyBiophysics and Systems BiologyRappaport Faculty of MedicineTechnion – Israel Institute of TechnologyHaifaIsrael
- Faculty of Biotechnology and Food EngineeringTechnion – Israel Institute of TechnologyHaifaIsrael
| | - Rita Shulman
- Department of PhysiologyBiophysics and Systems BiologyRappaport Faculty of MedicineTechnion – Israel Institute of TechnologyHaifaIsrael
| | - Irina Reiter
- Department of PhysiologyBiophysics and Systems BiologyRappaport Faculty of MedicineTechnion – Israel Institute of TechnologyHaifaIsrael
| | - Raz Palty
- Department of BiochemistryRappaport Faculty of MedicineTechnion – Israel Institute of TechnologyHaifaIsrael
| | - Michael Arad
- Leviev Heart CenterSheba Medical CenterRamat GanIsrael
- Sackler Faculty of MedicineTel Aviv UniversityTel AvivIsrael
| | - Eyal Gottlieb
- Department of Cell Biology and Cancer ScienceRappaport Faculty of MedicineTechnion – Israel Institute of TechnologyHaifaIsrael
| | - Ofer Binah
- Department of PhysiologyBiophysics and Systems BiologyRappaport Faculty of MedicineTechnion – Israel Institute of TechnologyHaifaIsrael
| |
Collapse
|
10
|
Ruiz M, Khairallah M, Dingar D, Vaniotis G, Khairallah RJ, Lauzier B, Thibault S, Trépanier J, Shi Y, Douillette A, Hussein B, Nawaito SA, Sahadevan P, Nguyen A, Sahmi F, Gillis MA, Sirois MG, Gaestel M, Stanley WC, Fiset C, Tardif JC, Allen BG. MK2-Deficient Mice Are Bradycardic and Display Delayed Hypertrophic Remodeling in Response to a Chronic Increase in Afterload. J Am Heart Assoc 2021; 10:e017791. [PMID: 33533257 PMCID: PMC7955338 DOI: 10.1161/jaha.120.017791] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Background Mitogen‐activated protein kinase–activated protein kinase‐2 (MK2) is a protein serine/threonine kinase activated by p38α/β. Herein, we examine the cardiac phenotype of pan MK2‐null (MK2−/−) mice. Methods and Results Survival curves for male MK2+/+ and MK2−/− mice did not differ (Mantel‐Cox test, P=0.580). At 12 weeks of age, MK2−/− mice exhibited normal systolic function along with signs of possible early diastolic dysfunction; however, aging was not associated with an abnormal reduction in diastolic function. Both R‐R interval and P‐R segment durations were prolonged in MK2‐deficient mice. However, heart rates normalized when isolated hearts were perfused ex vivo in working mode. Ca2+ transients evoked by field stimulation or caffeine were similar in ventricular myocytes from MK2+/+ and MK2−/− mice. MK2−/− mice had lower body temperature and an age‐dependent reduction in body weight. mRNA levels of key metabolic genes, including Ppargc1a, Acadm, Lipe, and Ucp3, were increased in hearts from MK2−/− mice. For equivalent respiration rates, mitochondria from MK2−/− hearts showed a significant decrease in Ca2+ sensitivity to mitochondrial permeability transition pore opening. Eight weeks of pressure overload increased left ventricular mass in MK2+/+ and MK2−/− mice; however, after 2 weeks the increase was significant in MK2+/+ but not MK2−/− mice. Finally, the pressure overload–induced decrease in systolic function was attenuated in MK2−/− mice 2 weeks, but not 8 weeks, after constriction of the transverse aorta. Conclusions Collectively, these results implicate MK2 in (1) autonomic regulation of heart rate, (2) cardiac mitochondrial function, and (3) the early stages of myocardial remodeling in response to chronic pressure overload.
Collapse
Affiliation(s)
- Matthieu Ruiz
- Department of Medicine Université de Montréal Québec Canada.,Montreal Heart Institute Montréal Québec Canada
| | - Maya Khairallah
- Department of Biochemistry and Molecular Medicine Université de Montréal Québec Canada.,Montreal Heart Institute Montréal Québec Canada
| | - Dharmendra Dingar
- Department of Biochemistry and Molecular Medicine Université de Montréal Québec Canada.,Montreal Heart Institute Montréal Québec Canada
| | - George Vaniotis
- Department of Biochemistry and Molecular Medicine Université de Montréal Québec Canada.,Montreal Heart Institute Montréal Québec Canada
| | | | | | - Simon Thibault
- Faculté de Pharmacie Université de Montréal Québec Canada.,Montreal Heart Institute Montréal Québec Canada
| | - Joëlle Trépanier
- Department of Biochemistry and Molecular Medicine Université de Montréal Québec Canada.,Montreal Heart Institute Montréal Québec Canada
| | - Yanfen Shi
- Montreal Heart Institute Montréal Québec Canada
| | | | | | - Sherin Ali Nawaito
- Department of Pharmacology and Physiology Université de Montréal Québec Canada.,Montreal Heart Institute Montréal Québec Canada.,Department of Physiology Faculty of Medicine Suez Canal University Ismailia Egypt
| | - Pramod Sahadevan
- Department of Biochemistry and Molecular Medicine Université de Montréal Québec Canada.,Montreal Heart Institute Montréal Québec Canada
| | - Albert Nguyen
- Department of Pharmacology and Physiology Université de Montréal Québec Canada.,Montreal Heart Institute Montréal Québec Canada
| | | | | | - Martin G Sirois
- Department of Pharmacology and Physiology Université de Montréal Québec Canada.,Montreal Heart Institute Montréal Québec Canada
| | - Matthias Gaestel
- Institute of Cell BiochemistryHannover Medical School Hannover Germany
| | | | - Céline Fiset
- Faculté de Pharmacie Université de Montréal Québec Canada.,Montreal Heart Institute Montréal Québec Canada
| | - Jean-Claude Tardif
- Department of Medicine Université de Montréal Québec Canada.,Montreal Heart Institute Montréal Québec Canada
| | - Bruce G Allen
- Department of Medicine Université de Montréal Québec Canada.,Department of Biochemistry and Molecular Medicine Université de Montréal Québec Canada.,Department of Pharmacology and Physiology Université de Montréal Québec Canada.,Montreal Heart Institute Montréal Québec Canada
| |
Collapse
|
11
|
Beamer E, Corrêa SAL. The p38 MAPK-MK2 Signaling Axis as a Critical Link Between Inflammation and Synaptic Transmission. Front Cell Dev Biol 2021; 9:635636. [PMID: 33585492 PMCID: PMC7876405 DOI: 10.3389/fcell.2021.635636] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/11/2021] [Indexed: 01/04/2023] Open
Abstract
p38 is a mitogen-activated protein kinase (MAPK), that responds primarily to stress stimuli. p38 has a number of targets for phosphorylation, including MAPK-activated protein kinase 2 (MK2). MK2 primarily functions as a master regulator of RNA-binding proteins, indirectly controlling gene expression at the level of translation. The role of MK2 in regulating the synthesis of pro-inflammatory cytokines downstream of inflammation and cellular stress is well-described. A significant amount of evidence, however, now points to a role for the p38MAPK-MK2 signaling axis in mediating synaptic plasticity through control of AMPA receptor trafficking and the morphology of dendritic spines. These processes are mediated through control of cytoskeletal dynamics via the activation of cofilin-1 and possibly control of the expression of Arc/Arg3.1. There is evidence that MK2 is necessary for group I metabotropic glutamate receptors long-term depression (mGluR-LTD). Disruption of this signaling may play an important role in mediating cognitive dysfunction in neurological disorders such as fragile X syndrome and Alzheimer’s disease. To date, the role of neuronal MK2 mediating synaptic plasticity in response to inflammatory stimuli has not yet been investigated. In immune cells, it is clear that MK2 is phosphorylated following activation of a broad range of cell surface receptors for cytokines and other inflammatory mediators. We propose that neuronal MK2 may be an important player in the link between inflammatory states and dysregulation of synaptic plasticity underlying cognitive functions. Finally, we discuss the potential of the p38MAPK-MK2 signaling axis as target for therapeutic intervention in a number of neurological disorders.
Collapse
Affiliation(s)
- Edward Beamer
- Faculty of Science and Engineering, Department of Life Sciences, Manchester Metropolitan University Manchester, Manchester, United Kingdom
| | - Sonia A L Corrêa
- Faculty of Science and Engineering, Department of Life Sciences, Manchester Metropolitan University Manchester, Manchester, United Kingdom
| |
Collapse
|
12
|
Pourbagher-Shahri AM, Farkhondeh T, Ashrafizadeh M, Talebi M, Samargahndian S. Curcumin and cardiovascular diseases: Focus on cellular targets and cascades. Biomed Pharmacother 2021; 136:111214. [PMID: 33450488 DOI: 10.1016/j.biopha.2020.111214] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 12/18/2020] [Accepted: 12/26/2020] [Indexed: 12/20/2022] Open
Abstract
Cardiovascular diseases (CVDs) are one of the leading causes of the most considerable mortality globally, and it has been tried to find the molecular mechanisms and design new drugs that triggered the molecular target. Curcumin is the main ingredient of Curcuma longa (turmeric) that has been used in traditional medicine for treating several diseases for years. Numerous investigations have indicated the beneficial effect of Curcumin in modulating multiple signaling pathways involved in oxidative stress, inflammation, apoptosis, and proliferation. The cardiovascular protective effects of Curcumin against CVDs have been indicated in several studies. In the current review study, we provided novel information on Curcumin's protective effects against various CVDs and potential molecular signaling targets of Curcumin. Nonetheless, more studies should be performed to discover the exact molecular target of Curcumin against CVDs.
Collapse
Affiliation(s)
| | - Tahereh Farkhondeh
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences (BUMS), Birjand, Iran; Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | - Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956 Istanbul, Turkey; Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956, Istanbul, Turkey
| | - Marjan Talebi
- Department of Pharmacognosy and Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, 19968 35115, Iran
| | - Saeed Samargahndian
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran.
| |
Collapse
|
13
|
p38 MAPK Pathway in the Heart: New Insights in Health and Disease. Int J Mol Sci 2020; 21:ijms21197412. [PMID: 33049962 PMCID: PMC7582802 DOI: 10.3390/ijms21197412] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/02/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023] Open
Abstract
The p38 mitogen-activated kinase (MAPK) family controls cell adaptation to stress stimuli. p38 function has been studied in depth in relation to cardiac development and function. The first isoform demonstrated to play an important role in cardiac development was p38α; however, all p38 family members are now known to collaborate in different aspects of cardiomyocyte differentiation and growth. p38 family members have been proposed to have protective and deleterious actions in the stressed myocardium, with the outcome of their action in part dependent on the model system under study and the identity of the activated p38 family member. Most studies to date have been performed with inhibitors that are not isoform-specific, and, consequently, knowledge remains very limited about how the different p38s control cardiac physiology and respond to cardiac stress. In this review, we summarize the current understanding of the role of the p38 pathway in cardiac physiology and discuss recent advances in the field.
Collapse
|
14
|
Stager M, Cheviron ZA. Is there a role for sarcolipin in avian facultative thermogenesis in extreme cold? Biol Lett 2020; 16:20200078. [PMID: 32516564 PMCID: PMC7336856 DOI: 10.1098/rsbl.2020.0078] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 04/27/2020] [Indexed: 11/12/2022] Open
Abstract
Endotherms defend their body temperature in the cold by employing shivering (ST) and/or non-shivering thermogenesis (NST). Although NST is well documented in mammals, its importance to avian heat generation is unclear. Recent work points to a prominent role for the sarco/endoplasmic reticulum Ca2+ ATPase (SERCA) in muscular NST. SERCA's involvement in both ST and NST, however, posits a tradeoff between these two heat-generating mechanisms. To explore this tradeoff, we assayed pectoralis gene expression of adult songbirds exposed to chronic temperature acclimations. Counter to mammal models, we found that cold-acclimated birds downregulated the expression of sarcolipin (SLN), a gene coding for a peptide that promotes heat generation by uncoupling SERCA Ca2+ transport from ATP hydrolysis, indicating a reduced potential for muscular NST. We also found differential expression of many genes involved in Ca2+ cycling and muscle contraction and propose that decreased SLN could promote increased pectoralis contractility for ST. Moreover, SLN transcript abundance negatively correlated with peak oxygen consumption under cold exposure (a proxy for ST) across individuals, and higher SLN transcript abundance escalated an individual's risk of hypothermia in acute cold. Our results therefore suggest that SLN-mediated NST may not be an important mechanism of-and could be a hindrance to-avian thermoregulation in extreme cold.
Collapse
Affiliation(s)
- Maria Stager
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
| | | |
Collapse
|
15
|
Mookerjee‐Basu J, Hooper R, Gross S, Schultz B, Go CK, Samakai E, Ladner J, Nicolas E, Tian Y, Zhou B, Zaidi MR, Tourtellotte W, He S, Zhang Y, Kappes DJ, Soboloff J. Suppression of Ca 2+ signals by EGR4 controls Th1 differentiation and anti-cancer immunity in vivo. EMBO Rep 2020; 21:e48904. [PMID: 32212315 PMCID: PMC7202224 DOI: 10.15252/embr.201948904] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 02/24/2020] [Accepted: 02/27/2020] [Indexed: 12/19/2022] Open
Abstract
While the zinc finger transcription factors EGR1, EGR2, and EGR3 are recognized as critical for T-cell function, the role of EGR4 remains unstudied. Here, we show that EGR4 is rapidly upregulated upon TCR engagement, serving as a critical "brake" on T-cell activation. Hence, TCR engagement of EGR4-/- T cells leads to enhanced Ca2+ responses, driving sustained NFAT activation and hyperproliferation. This causes profound increases in IFNγ production under resting and diverse polarizing conditions that could be reversed by pharmacological attenuation of Ca2+ entry. Finally, an in vivo melanoma lung colonization assay reveals enhanced anti-tumor immunity in EGR4-/- mice, attributable to Th1 bias, Treg loss, and increased CTL generation in the tumor microenvironment. Overall, these observations reveal for the first time that EGR4 is a key regulator of T-cell differentiation and function.
Collapse
Affiliation(s)
| | - Robert Hooper
- Fels Institute for Cancer Research and Molecular BiologyPhiladelphiaPAUSA,Department of Medical Genetics & Molecular BiochemistryTemple University School of MedicinePhiladelphiaPAUSA
| | - Scott Gross
- Fels Institute for Cancer Research and Molecular BiologyPhiladelphiaPAUSA,Department of Medical Genetics & Molecular BiochemistryTemple University School of MedicinePhiladelphiaPAUSA
| | - Bryant Schultz
- Fels Institute for Cancer Research and Molecular BiologyPhiladelphiaPAUSA,Department of Medical Genetics & Molecular BiochemistryTemple University School of MedicinePhiladelphiaPAUSA
| | - Christina K Go
- Fels Institute for Cancer Research and Molecular BiologyPhiladelphiaPAUSA,Department of Medical Genetics & Molecular BiochemistryTemple University School of MedicinePhiladelphiaPAUSA
| | - Elsie Samakai
- Fels Institute for Cancer Research and Molecular BiologyPhiladelphiaPAUSA,Department of Medical Genetics & Molecular BiochemistryTemple University School of MedicinePhiladelphiaPAUSA
| | | | | | - Yuanyuan Tian
- Fels Institute for Cancer Research and Molecular BiologyPhiladelphiaPAUSA,Department of ImmunologyTemple University School of MedicinePhiladelphiaPAUSA
| | - Bo Zhou
- Fels Institute for Cancer Research and Molecular BiologyPhiladelphiaPAUSA
| | - M Raza Zaidi
- Fels Institute for Cancer Research and Molecular BiologyPhiladelphiaPAUSA,Department of Medical Genetics & Molecular BiochemistryTemple University School of MedicinePhiladelphiaPAUSA
| | - Warren Tourtellotte
- Department of Pathology and Laboratory MedicineCedars Sinai Medical CenterWest HollywoodCAUSA
| | - Shan He
- Fels Institute for Cancer Research and Molecular BiologyPhiladelphiaPAUSA,Department of ImmunologyTemple University School of MedicinePhiladelphiaPAUSA
| | - Yi Zhang
- Fels Institute for Cancer Research and Molecular BiologyPhiladelphiaPAUSA,Department of ImmunologyTemple University School of MedicinePhiladelphiaPAUSA
| | | | - Jonathan Soboloff
- Fels Institute for Cancer Research and Molecular BiologyPhiladelphiaPAUSA,Department of Medical Genetics & Molecular BiochemistryTemple University School of MedicinePhiladelphiaPAUSA
| |
Collapse
|
16
|
What if? Mouse proteomics after gene inactivation. J Proteomics 2019; 199:102-122. [DOI: 10.1016/j.jprot.2019.03.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 03/09/2019] [Accepted: 03/10/2019] [Indexed: 12/17/2022]
|
17
|
Lv J, Deng C, Jiang S, Ji T, Yang Z, Wang Z, Yang Y. Blossoming 20: The Energetic Regulator's Birthday Unveils its Versatility in Cardiac Diseases. Am J Cancer Res 2019; 9:466-476. [PMID: 30809287 PMCID: PMC6376194 DOI: 10.7150/thno.29130] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 10/03/2018] [Indexed: 12/20/2022] Open
Abstract
The peroxisome proliferator-activated receptor γ (PPARγ) coactivator-1α (PGC-1α) was first identified in 1998 as a PGC-1 family member that regulates adaptive thermogenesis and mitochondrial function following cold exposure in brown adipose tissue. The PGC-1 family has drawn widespread attention over the past two decades as the energetic regulator. We recently summarized a review regarding PGC-1 signaling pathway and its mechanisms in cardiac metabolism. In this review, we elaborate upon the PGC-1 signaling network and highlight the recent progress of its versatile roles in cardiac diseases, including myocardial hypertrophy, peripartum and diabetic cardiomyopathy, and heart failure. The information reviewed here may be useful in future studies, which may increase the potential of this energetic regulator as a therapeutic target.
Collapse
|
18
|
EGR-mediated control of STIM expression and function. Cell Calcium 2018; 77:58-67. [PMID: 30553973 DOI: 10.1016/j.ceca.2018.12.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 12/03/2018] [Accepted: 12/04/2018] [Indexed: 12/22/2022]
Abstract
Ca2+ is a ubiquitous, dynamic and pluripotent second messenger with highly context-dependent roles in complex cellular processes such as differentiation, proliferation, and cell death. These Ca2+ signals are generated by Ca2+-permeable channels located on the plasma membrane (PM) and endoplasmic reticulum (ER) and shaped by PM- and ER-localized pumps and transporters. Differences in the expression of these Ca2+ homeostasis proteins contribute to cell and context-dependent differences in the spatiotemporal organization of Ca2+ signals and, ultimately, cell fate. This review focuses on the Early Growth Response (EGR) family of zinc finger transcription factors and their role in the transcriptional regulation of Stromal Interaction Molecule (STIM1), a critical regulator of Ca2+ entry in both excitable and non-excitable cells.
Collapse
|
19
|
Hsp70 Interacts with Mitogen-Activated Protein Kinase (MAPK)-Activated Protein Kinase 2 To Regulate p38MAPK Stability and Myoblast Differentiation during Skeletal Muscle Regeneration. Mol Cell Biol 2018; 38:MCB.00211-18. [PMID: 30275345 DOI: 10.1128/mcb.00211-18] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 09/26/2018] [Indexed: 12/24/2022] Open
Abstract
The regenerative process of injured muscle is dependent on the fusion and differentiation of myoblasts derived from muscle stem cells. Hsp70 is important for maintaining skeletal muscle homeostasis and regeneration, but the precise cellular mechanism remains elusive. In this study, we found that Hsp70 was upregulated during myoblast differentiation. Depletion or inhibition of Hsp70/Hsc70 impaired myoblast differentiation. Importantly, overexpression of p38 mitogen-activated protein kinase α (p38MAPKα) but not AKT1 rescued the impairment of myogenic differentiation in Hsp70- or Hsc70-depleted myoblasts. Moreover, Hsp70 interacted with MK2, a substrate of p38MAPK, to regulate the stability of p38MAPK. Knockdown of Hsp70 also led to downregulation of both MK2 and p38MAPK in intact muscles and during cardiotoxin-induced muscle regeneration. Hsp70 bound MK2 to regulate MK2-p38MAPK interaction in myoblasts. We subsequently identified the essential regions required for Hsp70-MK2 interaction. Functional analyses showed that MK2 is essential for both myoblast differentiation and skeletal muscle regeneration. Taken together, our findings reveal a novel role of Hsp70 in regulating myoblast differentiation by interacting with MK2 to stabilize p38MAPK.
Collapse
|
20
|
Cao Q, Zhang J, Gao L, Zhang Y, Dai M, Bao M. Dickkopf‑3 upregulation mediates the cardioprotective effects of curcumin on chronic heart failure. Mol Med Rep 2018; 17:7249-7257. [PMID: 29568962 PMCID: PMC5928680 DOI: 10.3892/mmr.2018.8783] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Accepted: 02/28/2018] [Indexed: 01/20/2023] Open
Abstract
Curcumin, isolated from rhizome of turmeric, has been widely studied as a potential therapeutic drug for cancer. However, protective effects of curcumin on chronic heart failure (CHF) have not been fully studied. In the present study, the effects of curcumin on CHF and the underlying mechanisms were investigated. A total of 40 rabbits were randomized into 4 groups: Control rabbits fed with placebo (Con) or curcumin (Con‑cur), CHF rabbits fed with placebo (CHF) or curcumin (CHF‑cur). CHF was induced by volume and pressure overload. The effects of curcumin on cardiac function and left ventricular (LV) structure were assessed by echocardiography and histology. The effects of curcumin on CHF molecular biomarkers were detected by dihydroethidium and immunohistochemical staining. The effects of curcumin on Dickkopf‑related protein 3 (DKK‑3), p38 mitogen‑activated protein kinase (p38), c‑Jun N‑terminal kinase (JNK) and apoptosis signal‑regulating kinase 1 (ASK1) were assessed by immunohistochemical staining and western blot analysis. Cardiac dysfunction and LV remodeling were successfully produced by ten weeks volume overload and eight weeks pressure overload in the CHF group. Compared with the Con group, the CHF group demonstrated higher levels of CHF molecular biomarkers, a lower level of DKK‑3 expression and alterations of p38, JNK and ASK1 protein expression. Curcumin alleviated all those abnormalities markedly in the CHF‑cur group. In summary, curcumin may exert cardioprotective effects by up‑regulating DKK‑3, which in turn may inhibit p38 and JNK signaling pathways in an ASK1‑dependent way. The present study demonstrated that Dickkopf‑3 upregulation mediates the cardioprotective effects of curcumin on chronic heart failure for the first time.
Collapse
Affiliation(s)
- Quan Cao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Junxia Zhang
- Department of Endocrinology, Wuhan General Hospital of the Chinese People's Liberation Army, Wuhan, Hubei 430070, P.R. China
| | - Ling Gao
- Department of Endocrinology and Metabolism, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yijie Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Mingyan Dai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Mingwei Bao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
21
|
Chang H, Jiang S, Ma X, Peng X, Zhang J, Wang Z, Xu S, Wang H, Gao Y. Proteomic analysis reveals the distinct energy and protein metabolism characteristics involved in myofiber type conversion and resistance of atrophy in the extensor digitorum longus muscle of hibernating Daurian ground squirrels. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2018; 26:20-31. [PMID: 29482114 DOI: 10.1016/j.cbd.2018.02.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 02/08/2018] [Accepted: 02/11/2018] [Indexed: 01/07/2023]
Abstract
Previous hibernation studies demonstrated that such a natural model of skeletal muscle disuse causes limited muscle atrophy and a significant fast-to-slow fiber type shift. However, the underlying mechanism as defined in a large-scale analysis remains unclarified. Isobaric tags for relative and absolute quantification (iTRAQ) based quantitative analysis were used to examine proteomic changes in the fast extensor digitorum longus muscles (EDL) of Daurian ground squirrels (Spermophilus dauricus). Although the wet weights and fiber cross-sectional area of the EDL muscle showed no significant decrease, the percentage of slow type fiber was 61% greater (P < 0.01) in the hibernation group. Proteomics analysis identified 264 proteins that were significantly changed (ratio < 0.83 or >1.2-fold and P < 0.05) in the hibernation group, of which 23 proteins were categorized into energy production and conversion and translation and 22 proteins were categorized into ribosomal structure and biogenesis. Along with the validation by western blot, MAPKAP kinase 2, ATP5D, ACADSB, calcineurin, CSTB and EIF2S were up-regulated in the hibernation group, whereas PDK4, COX II and EIF3C were down-regulated in the hibernation group. MAPKAP kinase 2 and PDK4 were associated with glycolysis, COX II and ATP5D were associated with oxidative phosphorylation, ACADSB was associated with fatty acid metabolism, calcineurin and CSTB were associated with catabolism, and EIF2S and EIF3C were associated with anabolism. Moreover, the total proteolysis rate of EDL in the hibernation group was significantly inhibited compared with that in the pre-hibernation group. These distinct energy and protein metabolism characteristics may be involved in myofiber type conversion and resistance to atrophy in the EDL of hibernating Daurian ground squirrels.
Collapse
Affiliation(s)
- Hui Chang
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an 710069, PR China; Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education, Xi'an 710069, PR China
| | - Shanfeng Jiang
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, PR China
| | - Xiufeng Ma
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an 710069, PR China
| | - Xin Peng
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an 710069, PR China
| | - Jie Zhang
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an 710069, PR China
| | - Zhe Wang
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an 710069, PR China
| | - Shenhui Xu
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an 710069, PR China
| | - Huiping Wang
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an 710069, PR China; Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education, Xi'an 710069, PR China
| | - Yunfang Gao
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an 710069, PR China; Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education, Xi'an 710069, PR China.
| |
Collapse
|
22
|
Yang Q, Li Y, Zhang X, Chen D. Zac1/GPR39 phosphorylating CaMK-II contributes to the distinct roles of Pax3 and Pax7 in myogenic progression. Biochim Biophys Acta Mol Basis Dis 2018; 1864:407-419. [DOI: 10.1016/j.bbadis.2017.10.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Revised: 09/15/2017] [Accepted: 10/22/2017] [Indexed: 12/12/2022]
|
23
|
O'Shea KM, Ananthakrishnan R, Li Q, Quadri N, Thiagarajan D, Sreejit G, Wang L, Zirpoli H, Aranda JF, Alberts AS, Schmidt AM, Ramasamy R. The Formin, DIAPH1, is a Key Modulator of Myocardial Ischemia/Reperfusion Injury. EBioMedicine 2017; 26:165-174. [PMID: 29239839 PMCID: PMC5832565 DOI: 10.1016/j.ebiom.2017.11.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 11/16/2017] [Accepted: 11/16/2017] [Indexed: 01/13/2023] Open
Abstract
The biochemical, ionic, and signaling changes that occur within cardiomyocytes subjected to ischemia are exacerbated by reperfusion; however, the precise mechanisms mediating myocardial ischemia/reperfusion (I/R) injury have not been fully elucidated. The receptor for advanced glycation end-products (RAGE) regulates the cellular response to cardiac tissue damage in I/R, an effect potentially mediated by the binding of the RAGE cytoplasmic domain to the diaphanous-related formin, DIAPH1. The aim of this study was to investigate the role of DIAPH1 in the physiological response to experimental myocardial I/R in mice. After subjecting wild-type mice to experimental I/R, myocardial DIAPH1 expression was increased, an effect that was echoed following hypoxia/reoxygenation (H/R) in H9C2 and AC16 cells. Further, compared to wild-type mice, genetic deletion of Diaph1 reduced infarct size and improved contractile function after I/R. Silencing Diaph1 in H9C2 cells subjected to H/R downregulated actin polymerization and serum response factor-regulated gene expression. Importantly, these changes led to increased expression of sarcoplasmic reticulum Ca2+ ATPase and reduced expression of the sodium calcium exchanger. This work demonstrates that DIAPH1 is required for the myocardial response to I/R, and that targeting DIAPH1 may represent an adjunctive approach for myocardial salvage after acute infarction.
Collapse
Affiliation(s)
- Karen M O'Shea
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, NY 10016, USA
| | - Radha Ananthakrishnan
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, NY 10016, USA
| | - Qing Li
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, NY 10016, USA
| | - Nosirudeen Quadri
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, NY 10016, USA
| | - Devi Thiagarajan
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, NY 10016, USA
| | - Gopalkrishna Sreejit
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, NY 10016, USA
| | - Lingjie Wang
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, NY 10016, USA
| | - Hylde Zirpoli
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, NY 10016, USA
| | - Juan Francisco Aranda
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, NY 10016, USA
| | - Arthur S Alberts
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Ann Marie Schmidt
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, NY 10016, USA
| | - Ravichandran Ramasamy
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, NY 10016, USA.
| |
Collapse
|
24
|
Ruiz M, Coderre L, Allen BG, Des Rosiers C. Protecting the heart through MK2 modulation, toward a role in diabetic cardiomyopathy and lipid metabolism. Biochim Biophys Acta Mol Basis Dis 2017; 1864:1914-1922. [PMID: 28735097 DOI: 10.1016/j.bbadis.2017.07.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 07/11/2017] [Accepted: 07/14/2017] [Indexed: 12/20/2022]
Abstract
Various signaling pathways have been identified in the heart as important players during development, physiological adaptation or pathological processes. This includes the MAPK families, particularly p38MAPK, which is involved in several key cellular processes, including differentiation, proliferation, apoptosis, inflammation, metabolism and survival. Disrupted p38MAPK signaling has been associated with several diseases, including cardiovascular diseases (CVD) as well as diabetes and its related complications. Despite efforts to translate this knowledge into therapeutic avenues, p38 inhibitors have failed in clinical trials due to adverse effects. Inhibition of MK2, a downstream target of p38, appears to be a promising alternative strategy. Targeting MK2 activity may avoid the adverse effects linked to p38 inhibition, while maintaining its beneficial effects. MK2 was first considered as a therapeutic target in inflammatory diseases such as rheumatoid polyarthritis. A growing body of evidence now supports a key role of MK2 signaling in the pathogenesis of CVD, particularly ischemia/reperfusion injury, hypertrophy, and hypertension and that its inhibition or inactivation is associated with improved heart and vascular functions. More recently, MK2 was shown to be a potential player in diabetes and related complications, particularly in liver and heart, and perturbations in calcium handling and lipid metabolism. In this review, we will discuss recent advances in our knowledge of the role of MK2 in p38MAPK-mediated signaling and the benefits of its loss of function in CVD and diabetes, with an emphasis on the roles of MK2 in calcium handling and lipid metabolism. This article is part of a Special issue entitled Cardiac adaptations to obesity, diabetes and insulin resistance, edited by Professors Jan F.C. Glatz, Jason R.B. Dyck and Christine Des Rosiers.
Collapse
Affiliation(s)
- Matthieu Ruiz
- Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada; Montreal Heart Institute, Research Center, 5000 Belanger Street, Montreal, Quebec, Canada
| | - Lise Coderre
- Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada; Department of Medicine, Université de Montréal, Montreal, Quebec, Canada; Montreal Heart Institute, Research Center, 5000 Belanger Street, Montreal, Quebec, Canada
| | - Bruce Gordon Allen
- Department of Biochemistry, Université de Montréal, Montreal, Quebec, Canada; Department of Medicine, Université de Montréal, Montreal, Quebec, Canada; Montreal Heart Institute, Research Center, 5000 Belanger Street, Montreal, Quebec, Canada.
| | - Christine Des Rosiers
- Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada; Department of Medicine, Université de Montréal, Montreal, Quebec, Canada; Montreal Heart Institute, Research Center, 5000 Belanger Street, Montreal, Quebec, Canada.
| |
Collapse
|
25
|
Grossi M, Bhattachariya A, Nordström I, Turczyńska KM, Svensson D, Albinsson S, Nilsson BO, Hellstrand P. Pyk2 inhibition promotes contractile differentiation in arterial smooth muscle. J Cell Physiol 2017; 232:3088-3102. [PMID: 28019664 DOI: 10.1002/jcp.25760] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 12/22/2016] [Accepted: 12/22/2016] [Indexed: 01/12/2023]
Abstract
Modulation from contractile to synthetic phenotype of vascular smooth muscle cells is a central process in disorders involving compromised integrity of the vascular wall. Phenotype modulation has been shown to include transition from voltage-dependent toward voltage-independent regulation of the intracellular calcium level, and inhibition of non-voltage dependent calcium influx contributes to maintenance of the contractile phenotype. One possible mediator of calcium-dependent signaling is the FAK-family non-receptor protein kinase Pyk2, which is activated by a number of stimuli in a calcium-dependent manner. We used the Pyk2 inhibitor PF-4594755 and Pyk2 siRNA to investigate the role of Pyk2 in phenotype modulation in rat carotid artery smooth muscle cells and in cultured intact arteries. Pyk2 inhibition promoted the expression of smooth muscle markers at the mRNA and protein levels under stimulation by FBS or PDGF-BB and counteracted phenotype shift in cultured intact carotid arteries and balloon injury ex vivo. During long-term (24-96 hr) treatment with PF-4594755, smooth muscle markers increased before cell proliferation was inhibited, correlating with decreased KLF4 expression and differing from effects of MEK inhibition. The Pyk2 inhibitor reduced Orai1 and preserved SERCA2a expression in carotid artery segments in organ culture, and eliminated the inhibitory effect of PDGF stimulation on L-type calcium channel and large-conductance calcium-activated potassium channel expression in carotid cells. Basal intracellular calcium level, calcium wave activity, and store-operated calcium influx were reduced after Pyk2 inhibition of growth-stimulated cells. Pyk2 inhibition may provide an interesting approach for preserving vascular smooth muscle differentiation under pathophysiological conditions.
Collapse
Affiliation(s)
- Mario Grossi
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | - Ina Nordström
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | - Daniel Svensson
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | - Bengt-Olof Nilsson
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Per Hellstrand
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
26
|
Acute low-intensity cycling with blood-flow restriction has no effect on metabolic signaling in human skeletal muscle compared to traditional exercise. Eur J Appl Physiol 2017; 117:345-358. [DOI: 10.1007/s00421-016-3530-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 12/29/2016] [Indexed: 10/20/2022]
|
27
|
Hu W, Xu T, Wu P, Pan D, Chen J, Chen J, Zhang B, Zhu H, Li D. Luteolin improves cardiac dysfunction in heart failure rats by regulating sarcoplasmic reticulum Ca 2+-ATPase 2a. Sci Rep 2017; 7:41017. [PMID: 28112209 PMCID: PMC5253630 DOI: 10.1038/srep41017] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 12/15/2016] [Indexed: 12/24/2022] Open
Abstract
We previously found that luteolin (Lut) appeared to improve the contractility of cardiomyocytes during ischemia/reperfusion in rats. The enhancement was associated with the alteration in sarcoplasmic reticulum Ca2+-ATPase 2a (SERCA2a). This finding prompted us to consider if the mechanism worked in heart failure (HF). We studied the regulation of SERCA2a by Lut in failing cardiomyocytes and intact heart of rats. Improvement of contractility and the mechanisms centered on SERCA2a were studied in isolated cardiomyocytes and intact heart. We found that Lut significantly improved contractility and Ca2+ transients, ameliorated expression, activity and stability of SERCA2a and upregulated expression of small ubiquitin-related modifier (SUMO) 1, which is a newfound SERCA2a regulator. Lut also increased phosphorylation of protein kinase B (Akt), phospholaban (PLB) and sumoylation of SERCA2a, specificity protein 1 (Sp1). Transcriptions of SUMO1 and SERCA2a were concurrently increased. Inhibition of posphatidylinositol 3 kinase/Akt (PI3K/Akt) pathway and SERCA2a activity both markedly abolished Lut-induced benefits in vitro and in vivo. Lut upregulated the expression ratio of Bcl-2/Bax, caspase-3/cleaved-Caspase3. Meanwhile, Lut ameliorated the myocardium fibrosis of HF. These discoveries provide an important potential therapeutic strategy that Lut targeted SERCA2a SUMOylation related to PI3K/Akt-mediated regulations on rescuing the dysfunction of HF.
Collapse
Affiliation(s)
- Wenjing Hu
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Tongda Xu
- Department of Cardiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China
| | - Pei Wu
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Defeng Pan
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Junhong Chen
- Department of Cardiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China
| | - Jing Chen
- Department of Cardiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China
| | - Buchun Zhang
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Hong Zhu
- Department of Cardiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China
| | - Dongye Li
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| |
Collapse
|
28
|
Jean-Charles A, Merle H, Audo I, Desoudin C, Bocquet B, Baudoin C, Sidibe M, Mauget-Faÿsse M, Wolff B, Fichard A, Lenaers G, Sahel JA, Gaudric A, Cohen SY, Hamel CP, Meunier I. Martinique Crinkled Retinal Pigment Epitheliopathy: Clinical Stages and Pathophysiologic Insights. Ophthalmology 2016; 123:2196-204. [PMID: 27474146 DOI: 10.1016/j.ophtha.2016.06.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 05/30/2016] [Accepted: 06/06/2016] [Indexed: 12/23/2022] Open
Abstract
PURPOSE To reappraise the autosomal dominant Martinique crinkled retinal pigment epitheliopathy (MCRPE) in light of the knowledge of its associated mutated gene mitogen-activated protein kinase-activated protein kinase 3 (MAPKAPK3), an actor in the p38 mitogen-activated protein kinase pathway. DESIGN Clinical and molecular study. PARTICIPANTS A total of 45 patients from 3 generations belonging to a family originating from Martinique with an autosomal dominant MCRPE were examined. METHODS Best-corrected visual acuity, fundus photographs, and spectral-domain optical coherence tomography (SD OCT) of all clinically affected patients and carriers for the causal mutation were reviewed at the initial visit and 4 years later for 10 of them. Histologic retinal lesions of Mapkapk3(-/-) mice were compared with those of the human disease. MAIN OUTCOME MEASURES The MCRPE natural history in view of MAPKAPK3 function and Mapkapk3(-/-) mouse retinal lesions. RESULTS Eighteen patients had the c.518T>C mutation. One heterozygous woman aged 20 years was asymptomatic with normal fundus and SD OCT (stage 0). All c.518T>C heterozygous patients older than 30 years of age had the characteristic dried-out soil fundus pattern (stages 1 and 2). Complications (stage 3) were observed in 7 cases, including polypoidal choroidal vasculopathy (PCV) and macular fibrosis or atrophy. One patient was homozygous and had a form with severe Bruch's membrane (BM) thickening and macular exudation with a dried-out soil pattern in the peripheral retina. The oldest heterozygous patient, who was legally blind, had peripheral nummular pigmentary changes (stage 4). After 4 years, visual acuity was unchanged in 6 of 10 patients. The dried-out soil elementary lesions radically enlarged in patients with a preferential macular extension and confluence. These findings are in line with the progressive thickening of BM noted with age in the mouse model. During follow-up, there was no occurrence of PCV. CONCLUSIONS MCRPE is an autosomal dominant, fully penetrant retinal dystrophy with a preclinical stage, an onset after the age of 30 years, and a preserved visual acuity until occurrence of macular complications. The natural history of MCRPE is in relation to the role of MAPKAPK3 in BM modeling, vascular endothelial growth factor activity, retinal pigment epithelial responses to aging, and oxidative stress.
Collapse
Affiliation(s)
- Albert Jean-Charles
- Department of Ophthalmology, University Hospital of Fort de France, Martinique (FWI), France
| | - Harold Merle
- Department of Ophthalmology, University Hospital of Fort de France, Martinique (FWI), France
| | - Isabelle Audo
- Fondation Adolphe de Rothschild, Paris, France; CHNO des Quinze-Vingts, DHU Sight Restore, INSERM-DHOS CIC1423, Paris - Sorborne Universités, UPMC Univ Paris 06, INSERM, CNRS, Institut de la Vision, Paris - Institute of Ophthalmology, University College of London, London, United Kingdom
| | | | - Béatrice Bocquet
- Institute for Neurosciences of Montpellier U1051, University of Montpellier - University Hospital, Genetics of Sensory Diseases, Montpellier, France
| | - Corinne Baudoin
- Institute for Neurosciences of Montpellier U1051, University of Montpellier - University Hospital, Genetics of Sensory Diseases, Montpellier, France
| | - Moro Sidibe
- Fondation Adolphe de Rothschild, Paris, France
| | | | - Benjamin Wolff
- Fondation Adolphe de Rothschild, Paris, France; Eye Clinic, Maison Rouge, Strasbourg, France
| | - Agnès Fichard
- Institute for Neurosciences of Montpellier U1051, University of Montpellier - University Hospital, Genetics of Sensory Diseases, Montpellier, France
| | - Guy Lenaers
- Institute for Neurosciences of Montpellier U1051, University of Montpellier - University Hospital, Genetics of Sensory Diseases, Montpellier, France
| | - José-Alain Sahel
- Fondation Adolphe de Rothschild, Paris, France; CHNO des Quinze-Vingts, DHU Sight Restore, INSERM-DHOS CIC1423, Paris - Sorborne Universités, UPMC Univ Paris 06, INSERM, CNRS, Institut de la Vision, Paris - Institute of Ophthalmology, University College of London, London, United Kingdom; Académie des Sciences, Institut de France, Paris, France
| | - Alain Gaudric
- Department of Ophthalmology, Lariboisière Hospital, Paris, France
| | - Salomon Yves Cohen
- Ophthalmic Center for Imaging and Laser, Paris, France; Department of Ophthalmology, Intercity Hospital and University Paris Est, Créteil, France
| | - Christian P Hamel
- Institute for Neurosciences of Montpellier U1051, University of Montpellier - University Hospital, Genetics of Sensory Diseases, Montpellier, France
| | - Isabelle Meunier
- Institute for Neurosciences of Montpellier U1051, University of Montpellier - University Hospital, Genetics of Sensory Diseases, Montpellier, France.
| |
Collapse
|
29
|
Derbré F, Droguet M, Léon K, Troadec S, Pennec JP, Giroux-Metges MA, Rannou F. Single Muscle Immobilization Decreases Single-Fibre Myosin Heavy Chain Polymorphism: Possible Involvement of p38 and JNK MAP Kinases. PLoS One 2016; 11:e0158630. [PMID: 27383612 PMCID: PMC4934689 DOI: 10.1371/journal.pone.0158630] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 06/20/2016] [Indexed: 11/29/2022] Open
Abstract
PURPOSE Muscle contractile phenotype is affected during immobilization. Myosin heavy chain (MHC) isoforms are the major determinant of the muscle contractile phenotype. We therefore sought to evaluate the effects of muscle immobilization on both the MHC composition at single-fibre level and the mitogen-activated protein kinases (MAPK), a family of intracellular signaling pathways involved in the stress-induced muscle plasticity. METHODS The distal tendon of female Wistar rat Peroneus Longus (PL) was cut and fixed to the adjacent bone at neutral muscle length. Four weeks after the surgery, immobilized and contralateral PL were dissociated and the isolated fibres were sampled to determine MHC composition. Protein kinase 38 (p38), extracellular signal-regulated kinases (ERK1/2), and c-Jun- NH2-terminal kinase (JNK) phosphorylations were measured in 6- and 15-day immobilized and contralateral PL. RESULTS MHC distribution in immobilized PL was as follows: I = 0%, IIa = 11.8 ± 2.8%, IIx = 53.0 ± 6.1%, IIb = 35.3 ± 7.3% and I = 6.1 ± 3.9%, IIa = 22.1 ± 3.4%, IIx = 46.6 ± 4.5%, IIb = 25.2 ± 6.6% in contralateral muscle. The MHC composition in immobilized muscle is consistent with a faster contractile phenotype according to the Hill's model of the force-velocity relationship. Immobilized and contralateral muscles displayed a polymorphism index of 31.1% (95% CI 26.1-36.0) and 39.3% (95% CI 37.0-41.5), respectively. Significant increases in p38 and JNK phosphorylation were observed following 6 and 15 days of immobilization. CONCLUSIONS Single muscle immobilization at neutral length induces a shift of MHC composition toward a faster contractile phenotype and decreases the polymorphic profile of single fibres. Activation of p38 and JNK could be a potential mechanism involved in these contractile phenotype modifications during muscle immobilization.
Collapse
Affiliation(s)
- Frédéric Derbré
- Laboratory “Movement Sport and health Sciences”(M2S) -EA1274, University Rennes 2-ENS Rennes, Rennes, France
| | - Mickaël Droguet
- Physiology Department-EA1274 M2S, School of Medicine, Brest, France
| | - Karelle Léon
- Physiology Department-EA1274 M2S, School of Medicine, Brest, France
| | - Samuel Troadec
- Physiology Department-EA1274 M2S, School of Medicine, Brest, France
| | | | | | - Fabrice Rannou
- Physiology Department-EA1274 M2S, School of Medicine, Brest, France
| |
Collapse
|
30
|
Wang S, Ding L, Ji H, Xu Z, Liu Q, Zheng Y. The Role of p38 MAPK in the Development of Diabetic Cardiomyopathy. Int J Mol Sci 2016; 17:ijms17071037. [PMID: 27376265 PMCID: PMC4964413 DOI: 10.3390/ijms17071037] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 06/20/2016] [Accepted: 06/24/2016] [Indexed: 02/06/2023] Open
Abstract
Diabetic cardiomyopathy (DCM) is a major complication of diabetes that contributes to an increase in mortality. A number of mechanisms potentially explain the development of DCM including oxidative stress, inflammation and extracellular fibrosis. Mitogen-activated protein kinase (MAPK)-mediated signaling pathways are common among these pathogenic responses. Among the diverse array of kinases, extensive attention has been given to p38 MAPK due to its capacity for promoting or inhibiting the translation of target genes. Growing evidence has indicated that p38 MAPK is aberrantly expressed in the cardiovascular system, including the heart, under both experimental and clinical diabetic conditions and, furthermore, inhibition of p38 MAPK activation in transgenic animal model or with its pharmacologic inhibitor significantly prevents the development of DCM, implicating p38 MAPK as a novel diagnostic indicator and therapeutic target for DCM. This review summarizes our current knowledge base to provide an overview of the impact of p38 MAPK signaling in diabetes-induced cardiac remodeling and dysfunction.
Collapse
Affiliation(s)
- Shudong Wang
- Cardiovascular Center, The First Hospital of Jilin University, Changchun 130021, China.
| | - Lijuan Ding
- Department of Radiation Oncology, the First Hospital of Jilin University, Changchun 130021, China.
| | - Honglei Ji
- Cardiovascular Center, The First Hospital of Jilin University, Changchun 130021, China.
| | - Zheng Xu
- Cardiovascular Center, The First Hospital of Jilin University, Changchun 130021, China.
| | - Quan Liu
- Cardiovascular Center, The First Hospital of Jilin University, Changchun 130021, China.
| | - Yang Zheng
- Cardiovascular Center, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
31
|
Wu HE, Baumgardt SL, Fang J, Paterson M, Liu Y, Du J, Shi Y, Qiao S, Bosnjak ZJ, Warltier DC, Kersten JR, Ge ZD. Cardiomyocyte GTP Cyclohydrolase 1 Protects the Heart Against Diabetic Cardiomyopathy. Sci Rep 2016; 6:27925. [PMID: 27295516 PMCID: PMC4904741 DOI: 10.1038/srep27925] [Citation(s) in RCA: 20] [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: 01/11/2016] [Accepted: 05/26/2016] [Indexed: 02/06/2023] Open
Abstract
Diabetic cardiomyopathy increases the risk of heart failure and death. At present, there are no effective approaches to preventing its development in the clinic. Here we report that reduction of cardiac GTP cyclohydrolase 1 (GCH1) degradation by genetic and pharmacological approaches protects the heart against diabetic cardiomyopathy. Diabetic cardiomyopathy was induced in C57BL/6 wild-type mice and transgenic mice with cardiomyocyte-specific overexpression of GCH1 with streptozotocin, and control animals were given citrate buffer. We found that diabetes-induced degradation of cardiac GCH1 proteins contributed to adverse cardiac remodeling and dysfunction in C57BL/6 mice, concomitant with decreases in tetrahydrobiopterin, dimeric and phosphorylated neuronal nitric oxide synthase, sarcoplasmic reticulum Ca(2+) handling proteins, intracellular [Ca(2+)]i, and sarcoplasmic reticulum Ca(2+) content and increases in phosphorylated p-38 mitogen-activated protein kinase and superoxide production. Interestingly, GCH-1 overexpression abrogated these detrimental effects of diabetes. Furthermore, we found that MG 132, an inhibitor for 26S proteasome, preserved cardiac GCH1 proteins and ameliorated cardiac remodeling and dysfunction during diabetes. This study deepens our understanding of impaired cardiac function in diabetes, identifies GCH1 as a modulator of cardiac remodeling and function, and reveals a new therapeutic target for diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Hsiang-En Wu
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
- National Institute on Drug Abuse, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MA 21224, USA
| | - Shelley L. Baumgardt
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Juan Fang
- Department of Pediatrics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Mark Paterson
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Yanan Liu
- Department of Medicine, Columbia University, 630 W. 168th Street, New York, NY 10032, USA
| | - Jianhai Du
- Department of Biochemistry, University of Washington, 1705 NE Pacific Street, Seattle, WA 98195, USA
| | - Yang Shi
- Aurora Research Institute, Aurora Health Care, 750 W. Virginia Street, Milwaukee, WI 53234, USA
| | - Shigang Qiao
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Zeljko J. Bosnjak
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - David C. Warltier
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Judy R. Kersten
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Zhi-Dong Ge
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| |
Collapse
|
32
|
Lodka D, Pahuja A, Geers-Knörr C, Scheibe RJ, Nowak M, Hamati J, Köhncke C, Purfürst B, Kanashova T, Schmidt S, Glass DJ, Morano I, Heuser A, Kraft T, Bassel-Duby R, Olson EN, Dittmar G, Sommer T, Fielitz J. Muscle RING-finger 2 and 3 maintain striated-muscle structure and function. J Cachexia Sarcopenia Muscle 2016; 7:165-80. [PMID: 27493870 PMCID: PMC4863828 DOI: 10.1002/jcsm.12057] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 05/24/2015] [Accepted: 06/04/2015] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND The Muscle-specific RING-finger (MuRF) protein family of E3 ubiquitin ligases is important for maintenance of muscular structure and function. MuRF proteins mediate adaptation of striated muscles to stress. MuRF2 and MuRF3 bind to microtubules and are implicated in sarcomere formation with noticeable functional redundancy. However, if this redundancy is important for muscle function in vivo is unknown. Our objective was to investigate cooperative function of MuRF2 and MuRF3 in the skeletal muscle and the heart in vivo. METHODS MuRF2 and MuRF3 double knockout mice (DKO) were generated and phenotypically characterized. Skeletal muscle and the heart were investigated by morphological measurements, histological analyses, electron microscopy, immunoblotting, and real-time PCR. Isolated muscles were subjected to in vitro force measurements. Cardiac function was determined by echocardiography and working heart preparations. Function of cardiomyocytes was measured in vitro. Cell culture experiments and mass-spectrometry were used for mechanistic analyses. RESULTS DKO mice showed a protein aggregate myopathy in skeletal muscle. Maximal force development was reduced in DKO soleus and extensor digitorum longus. Additionally, a fibre type shift towards slow/type I fibres occurred in DKO soleus and extensor digitorum longus. MuRF2 and MuRF3-deficient hearts showed decreased systolic and diastolic function. Further analyses revealed an increased expression of the myosin heavy chain isoform beta/slow and disturbed calcium handling as potential causes for the phenotype in DKO hearts. CONCLUSIONS The redundant function of MuRF2 and MuRF3 is important for maintenance of skeletal muscle and cardiac structure and function in vivo.
Collapse
Affiliation(s)
- Dörte Lodka
- Department of Molecular Cardiology, Experimental and Clinical Research Center (ECRC) Max Delbrück Center for Molecular Medicine and Charité Universitätsmedizin Berlin, Campus Buch 13125 Berlin Germany
| | - Aanchal Pahuja
- Institute of Molecular and Cell Physiology Hannover Medical School 30625 Hannover Germany
| | - Cornelia Geers-Knörr
- Institute of Molecular and Cell Physiology Hannover Medical School 30625 Hannover Germany
| | - Renate J Scheibe
- Institute of Physiological Chemistry Hannover Medical School 30625 Hannover Germany
| | - Marcel Nowak
- Department of Molecular Cardiology, Experimental and Clinical Research Center (ECRC) Max Delbrück Center for Molecular Medicine and Charité Universitätsmedizin Berlin, Campus Buch 13125 Berlin Germany; Department of Intracellular Proteolysis Max Delbrück Center for Molecular Medicine 13125 Berlin Germany
| | - Jida Hamati
- Department of Molecular Cardiology, Experimental and Clinical Research Center (ECRC) Max Delbrück Center for Molecular Medicine and Charité Universitätsmedizin Berlin, Campus Buch 13125 Berlin Germany
| | - Clemens Köhncke
- Department of Molecular Muscle Physiology Max Delbrück Center for Molecular Medicine 13125 Berlin Germany
| | - Bettina Purfürst
- Department of Electron Microscopy Max Delbrück Center for Molecular Medicine 13125 Berlin Germany
| | - Tamara Kanashova
- Department of Mass Spectrometry Max Delbrück Center for Molecular Medicine 13125 Berlin Germany
| | - Sibylle Schmidt
- Department of Molecular Cardiology, Experimental and Clinical Research Center (ECRC) Max Delbrück Center for Molecular Medicine and Charité Universitätsmedizin Berlin, Campus Buch 13125 Berlin Germany
| | - David J Glass
- Novartis Institutes for Biomedical Research Cambridge Massachusetts 02139 USA
| | - Ingo Morano
- Department of Molecular Muscle Physiology Max Delbrück Center for Molecular Medicine 13125 Berlin Germany
| | - Arnd Heuser
- Department of Cardiovascular Molecular Genetics Max Delbrück Center for Molecular Medicine 13125 Berlin Germany
| | - Theresia Kraft
- Institute of Molecular and Cell Physiology Hannover Medical School 30625 Hannover Germany
| | - Rhonda Bassel-Duby
- Department of Molecular Biology University of Texas Southwestern Medical Center Dallas Texas 75390-9148 USA
| | - Eric N Olson
- Department of Molecular Biology University of Texas Southwestern Medical Center Dallas Texas 75390-9148 USA
| | - Gunnar Dittmar
- Department of Mass Spectrometry Max Delbrück Center for Molecular Medicine 13125 Berlin Germany
| | - Thomas Sommer
- Department of Intracellular Proteolysis Max Delbrück Center for Molecular Medicine 13125 Berlin Germany
| | - Jens Fielitz
- Department of Molecular Cardiology, Experimental and Clinical Research Center (ECRC) Max Delbrück Center for Molecular Medicine and Charité Universitätsmedizin Berlin, Campus Buch 13125 Berlin Germany; Department of Cardiology Charité Universitätsmedizin Berlin, Campus Virchow 13353 Berlin Germany
| |
Collapse
|
33
|
Ruiz M, Coderre L, Lachance D, Houde V, Martel C, Thompson Legault J, Gillis MA, Bouchard B, Daneault C, Carpentier AC, Gaestel M, Allen BG, Des Rosiers C. MK2 Deletion in Mice Prevents Diabetes-Induced Perturbations in Lipid Metabolism and Cardiac Dysfunction. Diabetes 2016; 65:381-92. [PMID: 26558681 DOI: 10.2337/db15-0238] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 11/05/2015] [Indexed: 11/13/2022]
Abstract
Heart disease remains a major complication of diabetes, and the identification of new therapeutic targets is essential. This study investigates the role of the protein kinase MK2, a p38 mitogen-activated protein kinase downstream target, in the development of diabetes-induced cardiomyopathy. Diabetes was induced in control (MK2(+/+)) and MK2-null (MK2(-/-)) mice using repeated injections of a low dose of streptozotocin (STZ). This protocol generated in MK2(+/+) mice a model of diabetes characterized by a 50% decrease in plasma insulin, hyperglycemia, and insulin resistance (IR), as well as major contractile dysfunction, which was associated with alterations in proteins involved in calcium handling. While MK2(-/-)-STZ mice remained hyperglycemic, they showed improved IR and none of the cardiac functional or molecular alterations. Further analyses highlighted marked lipid perturbations in MK2(+/+)-STZ mice, which encompass increased 1) circulating levels of free fatty acid, ketone bodies, and long-chain acylcarnitines and 2) cardiac triglyceride accumulation and ex vivo palmitate β-oxidation. MK2(-/-)-STZ mice were also protected against all these diabetes-induced lipid alterations. Our results demonstrate the benefits of MK2 deletion on diabetes-induced cardiac molecular and lipid metabolic changes, as well as contractile dysfunction. As a result, MK2 represents a new potential therapeutic target to prevent diabetes-induced cardiac dysfunction.
Collapse
Affiliation(s)
- Matthieu Ruiz
- Department of Nutrition, Université de Montréal, Montréal, Québec, Canada Research Center, Montreal Heart Institute, Montréal, Québec, Canada
| | - Lise Coderre
- Department of Nutrition, Université de Montréal, Montréal, Québec, Canada Research Center, Montreal Heart Institute, Montréal, Québec, Canada Department of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Dominic Lachance
- Department of Nutrition, Université de Montréal, Montréal, Québec, Canada Research Center, Montreal Heart Institute, Montréal, Québec, Canada
| | - Valérie Houde
- Department of Nutrition, Université de Montréal, Montréal, Québec, Canada Research Center, Montreal Heart Institute, Montréal, Québec, Canada
| | - Cécile Martel
- Research Center, Montreal Heart Institute, Montréal, Québec, Canada
| | - Julie Thompson Legault
- Department of Nutrition, Université de Montréal, Montréal, Québec, Canada Research Center, Montreal Heart Institute, Montréal, Québec, Canada
| | | | - Bertrand Bouchard
- Department of Nutrition, Université de Montréal, Montréal, Québec, Canada Research Center, Montreal Heart Institute, Montréal, Québec, Canada
| | - Caroline Daneault
- Department of Nutrition, Université de Montréal, Montréal, Québec, Canada Research Center, Montreal Heart Institute, Montréal, Québec, Canada
| | - André C Carpentier
- Department of Medicine, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Matthias Gaestel
- Institute of Biochemistry, Hannover Medical School, Hannover, Germany
| | - Bruce G Allen
- Research Center, Montreal Heart Institute, Montréal, Québec, Canada Department of Medicine, Université de Montréal, Montréal, Québec, Canada Department of Biochemistry, Université de Montréal, Montréal, Québec, Canada
| | - Christine Des Rosiers
- Department of Nutrition, Université de Montréal, Montréal, Québec, Canada Research Center, Montreal Heart Institute, Montréal, Québec, Canada Department of Medicine, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
34
|
Kubis HP, Scheibe RJ, Decker B, Hufendiek K, Hanke N, Gros G, Meissner JD. Primary skeletal muscle cells cultured on gelatin bead microcarriers develop structural and biochemical features characteristic of adult skeletal muscle. Cell Biol Int 2016; 40:364-74. [PMID: 26610066 DOI: 10.1002/cbin.10565] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 11/23/2015] [Indexed: 12/15/2022]
Abstract
A primary skeletal muscle cell culture, in which myoblasts derived from newborn rabbit hindlimb muscles grow on gelatin bead microcarriers in suspension and differentiate into myotubes, has been established previously. In the course of differentiation and beginning spontaneous contractions, these multinucleated myotubes do not detach from their support. Here, we describe the development of the primary myotubes with respect to their ultrastructural differentiation. Scanning electron microscopy reveals that myotubes not only grow around the surface of one carrier bead but also attach themselves to neighboring carriers, forming bridges between carriers. Transmission electron microscopy demonstrates highly ordered myofibrils, T-tubules, and sarcoplasmic reticulum. The functionality of the contractile apparatus is evidenced by contractile activity that occurs spontaneously or can be elicited by electrostimulation. Creatine kinase activity increases steadily until day 20 of culture. Regarding the expression of isoforms of myosin heavy chains (MHC), we could demonstrate that from day 16 on, no non-adult MHC isoform mRNAs are present. Instead, on day 28 the myotubes express predominantly adult fast MHCIId/x mRNA and protein. This MHC pattern resembles that of fast muscles of adult rabbits. In contrast, primary myotubes grown on matrigel-covered culture dishes express substantial amounts of non-adult MHC protein even on day 21. To conclude, primary myotubes grown on microcarriers in their later stages exhibit many features of adult skeletal muscle and characteristics of fast type II fibers. Thus, the culture represents an excellent model of adult fast skeletal muscle, for example, when investigating molecular mechanisms of fast-to-slow fiber-type transformation.
Collapse
Affiliation(s)
- Hans-Peter Kubis
- Department of Physiology, Vegetative Physiology 4220, Hannover Medical School, 30625, Hannover, Germany
| | - Renate J Scheibe
- Department of Biochemistry, Institute of Physiological Chemistry, Hannover Medical School, 30625, Hannover, Germany
| | - Brigitte Decker
- Department of Anatomy, Institute of Cell Biology, Hannover Medical School, 30625, Hannover, Germany
| | - Karsten Hufendiek
- Department of Physiology, Vegetative Physiology 4220, Hannover Medical School, 30625, Hannover, Germany
| | - Nina Hanke
- Department of Physiology, Vegetative Physiology 4220, Hannover Medical School, 30625, Hannover, Germany
| | - Gerolf Gros
- Department of Physiology, Vegetative Physiology 4220, Hannover Medical School, 30625, Hannover, Germany
| | - Joachim D Meissner
- Department of Physiology, Vegetative Physiology 4220, Hannover Medical School, 30625, Hannover, Germany
| |
Collapse
|
35
|
Meunier I, Lenaers G, Bocquet B, Baudoin C, Piro-Megy C, Cubizolle A, Quilès M, Jean-Charles A, Cohen SY, Merle H, Gaudric A, Labesse G, Manes G, Péquignot M, Cazevieille C, Dhaenens CM, Fichard A, Ronkina N, Arthur SJ, Gaestel M, Hamel CP. A dominant mutation in MAPKAPK3, an actor of p38 signaling pathway, causes a new retinal dystrophy involving Bruch's membrane and retinal pigment epithelium. Hum Mol Genet 2016; 25:916-26. [PMID: 26744326 DOI: 10.1093/hmg/ddv624] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 12/21/2015] [Indexed: 01/09/2023] Open
Abstract
Inherited retinal dystrophies are clinically and genetically heterogeneous with significant number of cases remaining genetically unresolved. We studied a large family from the West Indies islands with a peculiar retinal disease, the Martinique crinkled retinal pigment epitheliopathy that begins around the age of 30 with retinal pigment epithelium (RPE) and Bruch's membrane changes resembling a dry desert land and ends with a retinitis pigmentosa. Whole-exome sequencing identified a heterozygous c.518T>C (p.Leu173Pro) mutation in MAPKAPK3 that segregates with the disease in 14 affected and 28 unaffected siblings from three generations. This unknown variant is predicted to be damaging by bioinformatic predictive tools and the mutated protein to be non-functional by crystal structure analysis. MAPKAPK3 is a serine/threonine protein kinase of the p38 signaling pathway that is activated by a variety of stress stimuli and is implicated in cellular responses and gene regulation. In contrast to other tissues, MAPKAPK3 is highly expressed in the RPE, suggesting a crucial role for retinal physiology. Expression of the mutated allele in HEK cells revealed a mislocalization of the protein in the cytoplasm, leading to cytoskeleton alteration and cytodieresis inhibition. In Mapkapk3-/- mice, Bruch's membrane is irregular with both abnormal thickened and thinned portions. In conclusion, we identified the first pathogenic mutation in MAPKAPK3 associated with a retinal disease. These findings shed new lights on Bruch's membrane/RPE pathophysiology and will open studies of this signaling pathway in diseases with RPE and Bruch's membrane alterations, such as age-related macular degeneration.
Collapse
Affiliation(s)
- Isabelle Meunier
- National Center in Genetic of Sensory Diseases, Institute for Neurosciences of Montpellier, INSERM U1051, University of Montpellier, Montpellier Hospital, Montpellier, France,
| | - Guy Lenaers
- National Center in Genetic of Sensory Diseases, Institute for Neurosciences of Montpellier, INSERM U1051, University of Montpellier, Montpellier Hospital, Montpellier, France, Mitochondrial Medicine Research Center, University of Angers, CNRS 6214, INSERM U1083, Angers, France
| | - Béatrice Bocquet
- National Center in Genetic of Sensory Diseases, Institute for Neurosciences of Montpellier, INSERM U1051, University of Montpellier, Montpellier Hospital, Montpellier, France
| | - Corinne Baudoin
- National Center in Genetic of Sensory Diseases, Institute for Neurosciences of Montpellier, INSERM U1051, University of Montpellier, Montpellier Hospital, Montpellier, France
| | - Camille Piro-Megy
- National Center in Genetic of Sensory Diseases, Institute for Neurosciences of Montpellier, INSERM U1051, University of Montpellier, Montpellier Hospital, Montpellier, France
| | - Aurélie Cubizolle
- National Center in Genetic of Sensory Diseases, Institute for Neurosciences of Montpellier, INSERM U1051, University of Montpellier, Montpellier Hospital, Montpellier, France
| | - Mélanie Quilès
- National Center in Genetic of Sensory Diseases, Institute for Neurosciences of Montpellier, INSERM U1051, University of Montpellier, Montpellier Hospital, Montpellier, France
| | - Albert Jean-Charles
- Department of Ophthalmology, University Hospital of Fort de France, Martinique (FWI), France
| | - Salomon Yves Cohen
- Imaging and Laser Center of Paris, Department of Ophthalmology, Intercity Hospital and University Paris, Creteil, France
| | - Harold Merle
- Department of Ophthalmology, University Hospital of Fort de France, Martinique (FWI), France
| | - Alain Gaudric
- Department of Ophthalmology, Lariboisiere Hospital, AP-HP and University Paris 7-Sorbonne Paris, Paris, France
| | - Gilles Labesse
- Center for Structural Biochemistry Montpellier, INSERM U1054-CNRS UMR5048, Montpellier, France
| | - Gaël Manes
- National Center in Genetic of Sensory Diseases, Institute for Neurosciences of Montpellier, INSERM U1051, University of Montpellier, Montpellier Hospital, Montpellier, France
| | - Marie Péquignot
- National Center in Genetic of Sensory Diseases, Institute for Neurosciences of Montpellier, INSERM U1051, University of Montpellier, Montpellier Hospital, Montpellier, France
| | - Chantal Cazevieille
- National Center in Genetic of Sensory Diseases, Institute for Neurosciences of Montpellier, INSERM U1051, University of Montpellier, Montpellier Hospital, Montpellier, France, Institute for Neurosciences, CRIC/IURC, Montpellier, France
| | - Claire-Marie Dhaenens
- CHRU Lille, Biochemistry and Molecular Biology Department, University Lille North, Lille, France
| | - Agnès Fichard
- National Center in Genetic of Sensory Diseases, Institute for Neurosciences of Montpellier, INSERM U1051, University of Montpellier, Montpellier Hospital, Montpellier, France
| | - Natalia Ronkina
- Institute of Biochemistry, Hannover Medical School, Hannover, Germany and
| | | | - Matthias Gaestel
- Institute of Biochemistry, Hannover Medical School, Hannover, Germany and
| | - Christian P Hamel
- National Center in Genetic of Sensory Diseases, Institute for Neurosciences of Montpellier, INSERM U1051, University of Montpellier, Montpellier Hospital, Montpellier, France
| |
Collapse
|
36
|
Abstract
Stress-response kinases, the mitogen-activated protein kinases (MAPKs) are activated in response to the challenge of a myriad of stressors. c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinases (ERKs), and p38 MAPKs are the predominant members of the MAPK family in the heart. Extensive studies have revealed critical roles of activated MAPKs in the processes of cardiac injury and heart failure and many other cardiovascular diseases. Recently, emerging evidence suggests that MAPKs also promote the development of cardiac arrhythmias. Thus, understanding the functional impact of MAPKs in the heart could shed new light on the development of novel therapeutic approaches to improve cardiac function and prevent arrhythmia development in the patients. This review will summarize the recent findings on the role of MAPKs in cardiac remodeling and arrhythmia development and point to the critical need of future studies to further elucidate the fundamental mechanisms of MAPK activation and arrhythmia development in the heart.
Collapse
|
37
|
Arabacilar P, Marber M. The case for inhibiting p38 mitogen-activated protein kinase in heart failure. Front Pharmacol 2015; 6:102. [PMID: 26029107 PMCID: PMC4428223 DOI: 10.3389/fphar.2015.00102] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 04/24/2015] [Indexed: 11/30/2022] Open
Abstract
This minireview discusses the evidence that the inhibition of p38 mitogen-activated protein kinases (p38 MAPKs) maybe of therapeutic value in heart failure. Most previous experimental studies, as well as past and ongoing clinical trials, have focussed on the role of p38 MAPKs in myocardial infarction and acute coronary syndromes. There is now growing evidence that these kinases are activated within the myocardium of the failing human heart and in the heart and blood vessels of animal models of heart failure. Furthermore, from a philosophical viewpoint the chronic activation of the adaptive stress pathways that lead to the activation of p38 MAPKs in heart failure is analogous to the chronic activation of the sympathetic, renin-aldosterone-angiotensin and neprilysin systems. These have provided some of the most effective therapies for heart failure. This minireview questions whether similar and synergistic advantages would follow the inhibition of p38 MAPKs.
Collapse
Affiliation(s)
- Pelin Arabacilar
- Cardiovascular Division, Department of Cardiology, King's College London British Heart Foundation Centre, The Rayne Institute, St Thomas' Hospital London, UK
| | - Michael Marber
- Cardiovascular Division, Department of Cardiology, King's College London British Heart Foundation Centre, The Rayne Institute, St Thomas' Hospital London, UK
| |
Collapse
|
38
|
Matsunaga Y, Qadota H, Furukawa M, Choe HH, Benian GM. Twitchin kinase interacts with MAPKAP kinase 2 in Caenorhabditis elegans striated muscle. Mol Biol Cell 2015; 26:2096-111. [PMID: 25851606 PMCID: PMC4472019 DOI: 10.1091/mbc.e14-05-1009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 04/01/2015] [Indexed: 01/13/2023] Open
Abstract
Titin-like giant polypeptides of muscle have protein kinase domains near their C-termini. These kinases are autoinhibited by portions of their own sequences. A putative activator for Caenorhabditis elegans twitchin kinase, MAK-1 (MAPKAP kinase 2), is expressed in nematode striated muscle, partially colocalizes with twitchin in sarcomeres, and binds to and phosphorylates twitchin kinase in vitro. In Caenorhabditis elegans, twitchin is a giant polypeptide located in muscle A-bands. The protein kinase of twitchin is autoinhibited by 45 residues upstream (NL) and 60 residues downstream (CRD) of the kinase catalytic core. Molecular dynamics simulation on a twitchin fragment revealed that the NL is released by pulling force. However, it is unclear how the CRD is removed. To identify proteins that may remove the CRD, we performed a yeast two-hybrid screen using twitchin kinase as bait. One interactor is MAK-1, C. elegans orthologue of MAPKAP kinase 2. MAPKAP kinase 2 is phosphorylated and activated by p38 MAP kinase. We demonstrate that the CRD of twitchin is important for binding to MAK-1. mak-1 is expressed in nematode body wall muscle, and antibodies to MAK-1 localize between and around Z-disk analogues and to the edge of A-bands. Whereas unc-22 mutants are completely resistant, mak-1 mutants are partially resistant to nicotine. MAK-1 can phosphorylate twitchin NL-Kin-CRD in vitro. Genetic data suggest the involvement of two other mak-1 paralogues and two orthologues of p38 MAP kinase. These results suggest that MAK-1 is an activator of twitchin kinase and that the p38 MAP kinase pathway may be involved in the regulation of twitchin.
Collapse
Affiliation(s)
- Yohei Matsunaga
- Department of Pathology, Emory University, Atlanta, GA 30322
| | - Hiroshi Qadota
- Department of Pathology, Emory University, Atlanta, GA 30322
| | - Miho Furukawa
- Department of Pathology, Emory University, Atlanta, GA 30322
| | | | - Guy M Benian
- Department of Pathology, Emory University, Atlanta, GA 30322
| |
Collapse
|
39
|
Ai X. SR calcium handling dysfunction, stress-response signaling pathways, and atrial fibrillation. Front Physiol 2015; 6:46. [PMID: 25745402 PMCID: PMC4333799 DOI: 10.3389/fphys.2015.00046] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 01/30/2015] [Indexed: 12/19/2022] Open
Abstract
Atrial fibrillation (AF) is the most common sustained arrhythmia. It is associated with a markedly increased risk of premature death due to embolic stroke and also complicates co-existing cardiovascular diseases such as heart failure. The prevalence of AF increases dramatically with age, and aging has been shown to be an independent risk of AF. Due to an aging population in the world, a growing body of AF patients are suffering a diminished quality of life and causing an associated economic burden. However, effective pharmacologic treatments and prevention strategies are lacking due to a poor understanding of the molecular and electrophysiologic mechanisms of AF in the failing and/or aged heart. Recent studies suggest that altered atrial calcium handling contributes to the onset and maintenance of AF. Here we review the role of stress-response kinases and calcium handling dysfunction in AF genesis in the aged and failing heart.
Collapse
Affiliation(s)
- Xun Ai
- Department of Cell and Molecular Physiology, Loyola University Chicago Maywood, IL, USA
| |
Collapse
|
40
|
Wernbom M, Apro W, Paulsen G, Nilsen TS, Blomstrand E, Raastad T. Acute low-load resistance exercise with and without blood flow restriction increased protein signalling and number of satellite cells in human skeletal muscle. Eur J Appl Physiol 2014; 113:2953-65. [PMID: 24078212 DOI: 10.1007/s00421-013-2733-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Accepted: 09/16/2013] [Indexed: 11/28/2022]
Abstract
PURPOSE To investigate hypertrophic signalling after a single bout of low-load resistance exercise with and without blood flow restriction (BFR). METHODS Seven subjects performed unilateral knee extensions at 30 % of their one repetition maximum. The subjects performed five sets to failure with BFR on one leg, and then repeated the same amount of work with the other leg without BFR. Biopsies were obtained from m. vastus lateralis before and 1, 24 and 48 h after exercise. RESULTS At 1-h post-exercise, phosphorylation of p70S6KThr389 and p38MAPKThr180/Tyr182 was elevated in the BFR leg, but not in the free-flow leg. Phospho-p70S6KThr389 was elevated three- to fourfold in both legs at 24-h post-exercise, but back to baseline at 48 h. The number of visible satellite cells (SCs) per muscle fibre was increased for all post-exercise time points and in both legs (33–53 %). The proportion of SCs with cytoplasmic extensions was elevated at 1-h post in the BFR leg and the number of SCs positive for myogenin and/or MyoD was increased at 1- and 24-h post-exercise for both legs combined. CONCLUSION Acute low-load resistance exercise with BFR resulted in early (1 h) and late (24 h) enhancement of phospho-p70S6KThr389, an early response of p38MAPK, and an increased number of SCs per muscle fibre. Enhanced phospho-p70S6KThr389 at 24-h post-exercise and increases in SC numbers were seen also in the free-flow leg. Implications of these findings for the hypertrophic effects of fatiguing low-load resistance exercise with and without BFR are discussed.
Collapse
|