1
|
Hao S, Lasaracina AP, Epps J, Ferreri NR. TNF inhibits NKCC2 phosphorylation by a calcineurin-dependent pathway. Am J Physiol Renal Physiol 2025; 328:F489-F500. [PMID: 40062390 PMCID: PMC12048884 DOI: 10.1152/ajprenal.00251.2024] [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: 08/26/2024] [Revised: 09/30/2024] [Accepted: 02/26/2025] [Indexed: 03/20/2025] Open
Abstract
We previously demonstrated that tumor necrosis factor-alpha (TNF) inhibits Na+-K+-2Cl- cotransporter (NKCC2) phosphorylation in the thick ascending limb (TAL); however, the underlying mechanism remains unclear. We tested the hypothesis that the induction of calcineurin (CN) activity and the expression of CN isoforms contribute to the mechanism by which TNF inhibits phospho-NKCC2 (pNKCC2) expression. CN activity increased by approximately twofold in primary cultures of medullary (m)TAL cells challenged with mouse recombinant TNF. In contrast, silencing TNF production in mTAL cells using lentivirus U6-TNF-ex4 reduced CN activity. pNKCC2 expression decreased in mTAL cells challenged with TNF, whereas inhibition of CN activity with cyclosporine A (CsA) increased pNKCC2 expression. Although mTAL cells express both the calcineurin A subunit (CNA) α and β isoforms, only CNA β isoform mRNA increased after mTAL cells were challenged with TNF. In vivo, both TNF and CNA β expression increased in outer medulla (OM) from mice given 1% NaCl in the drinking water for 7 days and intrarenal lentivirus silencing of TNF selectively reduced expression of CNA β. Intrarenal injection of a lentivirus that specifically silenced CNA β (U6-CNAβ-ex6) increased pNKCC2 expression and attenuated the inhibitory effects of TNF on pNKCC2 expression in freshly isolated TAL tubules. Collectively, the study is the first to demonstrate that TNF increases CN activity and specifically induces β-isoform expression in the kidney. Since NKCC2 is a known target of the CNA β isoform, these findings suggest that a CN-dependent signaling pathway involving this isoform contributes to the mechanism by which TNF inhibits pNKCC2 expression.NEW & NOTEWORTHY The beneficial immunosuppressive effects of CsA are tempered by renal side effects including reduction of GFR, proximal tubule damage, reduced urinary concentration, fibrosis and hypertension. As chronic administration of CN inhibitors frequently induce hypertension and renal nephropathy in humans, understanding the molecular mechanisms by which CN isoforms regulate the activity of renal transporters may provide the framework for developing new drugs that more selectively modulate the diverse functions of CN.
Collapse
Affiliation(s)
- Shoujin Hao
- Department of Pharmacology, New York Medical College, Valhalla, New York, United States
| | - Anna Pia Lasaracina
- Department of Pharmacology, New York Medical College, Valhalla, New York, United States
| | - Jarred Epps
- Department of Pharmacology, New York Medical College, Valhalla, New York, United States
| | - Nicholas R Ferreri
- Department of Pharmacology, New York Medical College, Valhalla, New York, United States
| |
Collapse
|
2
|
Xia Q, Liu X, Zhong L, Qu J, Dong L. SMURF1 mediates damaged lysosomal homeostasis by ubiquitinating PPP3CB to promote the activation of TFEB. Autophagy 2025; 21:530-547. [PMID: 39324484 PMCID: PMC11849922 DOI: 10.1080/15548627.2024.2407709] [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: 11/27/2023] [Revised: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024] Open
Abstract
The calcium-activated phosphatase PPP3/calcineurin dephosphorylates TFEB (transcription factor EB) to trigger its nuclear translocation and the activation of macroautophagic/autophagic targets. However, the detailed molecular mechanism regulating TFEB activation remains poorly understood. Here, we highlighted the importance of SMURF1 (SMAD specific E3 ubiquitin protein ligase 1) in the activation of TFEB for lysosomal homeostasis. SMURF1 deficiency prevents the calcium-triggered ubiquitination of the catalytic subunit of PPP3/calcineurin in a manner consistent with defective autophagic degradation of damaged lysosomes. Mechanically, PPP3CB/CNA2 plays a bridging role in the recruitment of SMURF1 by LGALS3 (galectin 3) upon lysosome damage. Importantly, PPP3CB increases the dissociation of the N-terminal tail (NT) and C-terminal carbohydrate-recognition domain (CRD) of LGALS3, which may promote the formation of open conformers in a PPP3CB dephosphorylation activity-dependent manner. In addition, PPP3CB is ubiquitinated at lysine 146 by the recruited SMURF1 in response to intracellular calcium stimulation. The K63-linked ubiquitination of PPP3CB enhances the recruitment of TFEB. Moreover, TFEB directly interacts with both PPP3CB and the regulatory subunit PPP3R1 which facilitate the conformational correction of TFEB for its activation for the transcription of TFEB-targeted genes. Altogether, our results highlighted a critical mechanism for the regulation of PPP3/calcineurin activity via its ubiquitin ligase SMURF1 in response to lysosomal membrane damage, which may account for a potential target for the treatment of stress-related diseases.Abbreviation AID: autoinhibitory domain; ATG: autophagy related; CD: catalytic domain; CRD: carbohydrate-recognition domain; CsA: cyclosporin A; DMSO: dimethyl sulfoxide; ESCRT: endosomal sorting complexes required for transport; GSK3B: glycogen synthase kinase 3 beta; LAMP1: lysosomal associated membrane protein 1; LGALS3: galectin 3; LLOMe: L-leucyl-L-leucine methyl ester; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; ML-SA1: mucolipin synthetic agonist 1; MTORC1: mechanistic target of rapamycin kinase complex 1; NT: N-terminal tail; PPP3CB: protein phosphatase 3 catalytic subunit beta; PPP3R1: protein phosphatase 3 regulatory subunit B, alpha; SMURF1: SMAD specific E3 ubiquitin protein ligase 1; SQSTM1/p62: sequestosome 1; TFEB: transcription factor EB; VCP/p97: valosin containing protein; YWHA/14-3-3: tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein.
Collapse
Affiliation(s)
- Qin Xia
- Department of General Surgery, Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, China
- State Key Laboratory of Hearing and Balance Science and Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Xuan Liu
- State Key Laboratory of Hearing and Balance Science and Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Lu Zhong
- State Key Laboratory of Hearing and Balance Science and Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Jun Qu
- Department of General Surgery, Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Lei Dong
- Department of General Surgery, Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, China
- State Key Laboratory of Hearing and Balance Science and Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), School of Life Science, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
3
|
Salagre D, Bajit H, Fernández-Vázquez G, Dwairy M, Garzón I, Haro-López R, Agil A. Melatonin induces fiber switching by improvement of mitochondrial oxidative capacity and function via NRF2/RCAN/MEF2 in the vastus lateralis muscle from both sex Zücker diabetic fatty rats. Free Radic Biol Med 2025; 227:322-335. [PMID: 39645208 DOI: 10.1016/j.freeradbiomed.2024.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 11/19/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
The positive role of melatonin in obesity control and skeletal muscle (SKM) preservation is well known. We recently showed that melatonin improves vastus lateralis muscle (VL) fiber oxidative phenotype. However, fiber type characterization, mitochondrial function, and molecular mechanisms that underlie VL fiber switching by melatonin are still undefined. Our study aims to investigate whether melatonin induces fiber switching by NRF2/RCAN/MEF2 pathway activation and mitochondrial oxidative metabolism modulation in the VL of both sex Zücker diabetic fatty (ZDF) rats. 5-Weeks-old male and female ZDF rats (N = 16) and their age-matched lean littermates (ZL) were subdivided into two subgroups: control (C) and orally treated with melatonin (M) (10 mg/kg/day) for 12 weeks. Interestingly, melatonin increased oxidative fibers amounts (Types I and IIa) counteracting the decreased levels found in the VL of obese-diabetic rats, and upregulated NRF2, calcineurin and MEF2 expression. Melatonin also restored the mitochondrial oxidative capacity increasing the respiratory control ratio (RCR) in both sex and phenotype rats through the reduction of the proton leak component of respiration (state 4). Melatonin also improved the VL mitochondrial phosphorylation coefficient and modulated the total oxygen consumption by enhancing complex I, III and IV activity, and fatty acid oxidation (FAO) in both sex obese-diabetic rats, decreasing in male and increasing in female the complex II oxygen consumption. These findings suggest that melatonin treatment induces fiber switching in SKM improving mitochondrial functionality by NRF2/RCAN/MEF2 pathway activation.
Collapse
Affiliation(s)
- Diego Salagre
- Department of Pharmacology, BioHealth Institute Granada (IBs Granada), Neuroscience Institute (CIBM), School of Medicine, University of Granada, 18016, Granada, Spain
| | - Habiba Bajit
- Department of Pharmacology, BioHealth Institute Granada (IBs Granada), Neuroscience Institute (CIBM), School of Medicine, University of Granada, 18016, Granada, Spain
| | | | - Mutaz Dwairy
- Department of Civil Engineering, Yarmuk University, 21163, Irbid, Jordan
| | - Ingrid Garzón
- Tissue Engineering Group, Department of Histology, BioHealth Institute Granada (IBs Granada), School of Medicine, University of Granada, 18016, Granada, Spain
| | - Rocío Haro-López
- Department of Pharmacology, BioHealth Institute Granada (IBs Granada), Neuroscience Institute (CIBM), School of Medicine, University of Granada, 18016, Granada, Spain
| | - Ahmad Agil
- Department of Pharmacology, BioHealth Institute Granada (IBs Granada), Neuroscience Institute (CIBM), School of Medicine, University of Granada, 18016, Granada, Spain.
| |
Collapse
|
4
|
Das P, Aballay A, Singh J. Calcineurin inhibition enhances Caenorhabditis elegans lifespan by defecation defects-mediated calorie restriction and nuclear hormone signaling. eLife 2024; 12:RP89572. [PMID: 39485281 PMCID: PMC11530235 DOI: 10.7554/elife.89572] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024] Open
Abstract
Calcineurin is a highly conserved calcium/calmodulin-dependent serine/threonine protein phosphatase with diverse functions. Inhibition of calcineurin is known to enhance the lifespan of Caenorhabditis elegans through multiple signaling pathways. Aiming to study the role of calcineurin in regulating innate immunity, we discover that calcineurin is required for the rhythmic defecation motor program (DMP) in C. elegans. Calcineurin inhibition leads to defects in the DMP, resulting in intestinal bloating, rapid colonization of the gut by bacteria, and increased susceptibility to bacterial infection. We demonstrate that intestinal bloating caused by calcineurin inhibition mimics the effects of calorie restriction, resulting in enhanced lifespan. The TFEB ortholog, HLH-30, is required for lifespan extension mediated by calcineurin inhibition. Finally, we show that the nuclear hormone receptor, NHR-8, is upregulated by calcineurin inhibition and is necessary for the increased lifespan. Our studies uncover a role for calcineurin in the C. elegans DMP and provide a new mechanism for calcineurin inhibition-mediated longevity extension.
Collapse
Affiliation(s)
- Priyanka Das
- Department of Biological Sciences, Indian Institute of Science Education and ResearchMohaliIndia
| | - Alejandro Aballay
- Department of Genetics, The University of Texas MD Anderson Cancer CenterHoustonUnited States
| | - Jogender Singh
- Department of Biological Sciences, Indian Institute of Science Education and ResearchMohaliIndia
| |
Collapse
|
5
|
Wass SY, Sun H, Tchou G, Liu N, Van Wagoner DR, Chung MK, Barnard J, Smith JD. Transcriptomic Insights into the Atrial Fibrillation Susceptibility Locus near the MYOZ1 and SYNPO2L Genes. Int J Mol Sci 2024; 25:10309. [PMID: 39408638 PMCID: PMC11477451 DOI: 10.3390/ijms251910309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/15/2024] [Accepted: 09/19/2024] [Indexed: 10/20/2024] Open
Abstract
Genome-wide association studies have identified a locus on chromosome 10q22, where many co-inherited single nucleotide polymorphisms (SNPs) are associated with atrial fibrillation (AF). This study seeks to identify the impact of this locus on gene expression at the transcript isoform level in human left atria and to gain insight into potential causal variants. Bulk RNA sequencing was analyzed to identify myozenin 1 (MYOZ1) and synaptopodin 2-like (SYNPO2L) transcript isoforms and the association of common SNPs in this region with transcript isoform expression levels. Chromatin marks were used to suggest candidate regulatory SNPs in this region. Protein amino acid changes were examined for predicted functional consequences. Transfection of MYOZ1 and two SYNPO2L isoforms were performed to localize their encoded proteins in cardiomyocytes derived from stem cells. We identified one MYOZ1 transcript isoform and four SYNPO2L transcript isoforms, two of which encode proteins, while the other two encode long noncoding RNAs (lncRNAs). The risk allele of the strongest AF susceptibility SNP on chromosome 10q22 is associated with decreased MYOZ1 expression and increased expression of the two SNYPO2L lncRNA isoforms. There are many SNPs co-inherited with the top AF-associated SNP due to linkage disequilibrium (LD), including rs11000728, which we propose as the MYOZ1 regulatory SNP, confirmed by reporter gene transfection. In addition, this LD block includes three missense SNPs in the SYNPO2L gene, with the minor protective haplotype predicted to be detrimental to protein function. MYOZ1 and both protein isoforms of SYNPO2L were localized to the sarcomere. This is a complex locus with the potential for several SNPs in a haplotype to alter AF susceptibility by opposing effects on MYOZ1 and SYNPO2L lncRNA expression, along with effects on SYNPO2L protein function.
Collapse
Affiliation(s)
- Sojin Y. Wass
- Departments of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Han Sun
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Gregory Tchou
- Departments of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Nana Liu
- Departments of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, OH 44195, USA
| | - David R. Van Wagoner
- Departments of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Mina K. Chung
- Departments of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH 44195, USA
| | - John Barnard
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jonathan D. Smith
- Departments of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
| |
Collapse
|
6
|
Padilha SF, Ibelli AMG, Peixoto JO, Cantão ME, Moreira GCM, Fernandes LT, Tavernari FC, Morés MAZ, Bastos APA, Dias LT, Teixeira RA, Ledur MC. Novel Candidate Genes Involved in an Initial Stage of White Striping Development in Broiler Chickens. Animals (Basel) 2024; 14:2379. [PMID: 39199913 PMCID: PMC11350825 DOI: 10.3390/ani14162379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 09/01/2024] Open
Abstract
White striping (WS) is a myopathy characterized by the appearance of white stripes parallel to the muscle fibers in the breast of broiler chickens, composed of adipose and connective tissues. This condition causes economic losses and, although common, its etiology remains poorly understood. Hence, the objective was to identify genes and biological mechanisms involved in the early stages of WS using a paternal broiler line that grows slightly slower than commercial ones, at 35 days of age, through the RNA sequencing of the pectoralis major muscle. Thirty genes were differentially expressed between normal and WS-affected chickens, with 23 upregulated and 7 downregulated in the affected broilers. Of these, 14 genes are novel candidates for WS and are implicated in biological processes related to muscle development (CEPBD, DUSP8, METTL21EP, NELL2, and UBE3D), lipid metabolism (PDK4, DDIT4, FKBP5, DGAT2, LIPG, TDH, and RGCC), and collagen (COL4A5 and COL4A6). Genes related to changes in muscle fiber type and the processes of apoptosis, autophagy, proliferation, and differentiation are possibly involved with the initial stage of WS development. In contrast, the genes linked to lipid metabolism and collagen may have their expression altered due to the progression of the myopathy.
Collapse
Affiliation(s)
- Suelen Fernandes Padilha
- Departamento de Zootecnia, Programa de Pós-Graduação em Zootecnia, Universidade Federal do Paraná, Curitiba 80035-050, PR, Brazil; (S.F.P.); (L.T.D.); (R.A.T.)
| | - Adriana Mércia Guaratini Ibelli
- Embrapa Suínos e Aves, Concórdia 89715-899, SC, Brazil; (J.O.P.); (M.E.C.); (L.T.F.); (F.C.T.); (M.A.Z.M.); (A.P.A.B.)
- Programa de Pós-Graduação em Ciências Veterinárias, Universidade Estadual do Centro Oeste, Guarapuava 85040-167, PR, Brazil
| | - Jane Oliveira Peixoto
- Embrapa Suínos e Aves, Concórdia 89715-899, SC, Brazil; (J.O.P.); (M.E.C.); (L.T.F.); (F.C.T.); (M.A.Z.M.); (A.P.A.B.)
- Programa de Pós-Graduação em Ciências Veterinárias, Universidade Estadual do Centro Oeste, Guarapuava 85040-167, PR, Brazil
| | - Maurício Egídio Cantão
- Embrapa Suínos e Aves, Concórdia 89715-899, SC, Brazil; (J.O.P.); (M.E.C.); (L.T.F.); (F.C.T.); (M.A.Z.M.); (A.P.A.B.)
| | | | - Lana Teixeira Fernandes
- Embrapa Suínos e Aves, Concórdia 89715-899, SC, Brazil; (J.O.P.); (M.E.C.); (L.T.F.); (F.C.T.); (M.A.Z.M.); (A.P.A.B.)
| | - Fernando Castro Tavernari
- Embrapa Suínos e Aves, Concórdia 89715-899, SC, Brazil; (J.O.P.); (M.E.C.); (L.T.F.); (F.C.T.); (M.A.Z.M.); (A.P.A.B.)
- Programa de Pós-Graduação em Zootecnia, Universidade do Estado de Santa Catarina, UDESC-Oeste, Chapecó 89815-630, SC, Brazil
| | - Marcos Antônio Zanella Morés
- Embrapa Suínos e Aves, Concórdia 89715-899, SC, Brazil; (J.O.P.); (M.E.C.); (L.T.F.); (F.C.T.); (M.A.Z.M.); (A.P.A.B.)
| | - Ana Paula Almeida Bastos
- Embrapa Suínos e Aves, Concórdia 89715-899, SC, Brazil; (J.O.P.); (M.E.C.); (L.T.F.); (F.C.T.); (M.A.Z.M.); (A.P.A.B.)
- Programa de Pós-Graduação em Ciências Veterinárias, Universidade Estadual do Centro Oeste, Guarapuava 85040-167, PR, Brazil
| | - Laila Talarico Dias
- Departamento de Zootecnia, Programa de Pós-Graduação em Zootecnia, Universidade Federal do Paraná, Curitiba 80035-050, PR, Brazil; (S.F.P.); (L.T.D.); (R.A.T.)
| | - Rodrigo Almeida Teixeira
- Departamento de Zootecnia, Programa de Pós-Graduação em Zootecnia, Universidade Federal do Paraná, Curitiba 80035-050, PR, Brazil; (S.F.P.); (L.T.D.); (R.A.T.)
| | - Mônica Corrêa Ledur
- Embrapa Suínos e Aves, Concórdia 89715-899, SC, Brazil; (J.O.P.); (M.E.C.); (L.T.F.); (F.C.T.); (M.A.Z.M.); (A.P.A.B.)
- Programa de Pós-Graduação em Zootecnia, Universidade do Estado de Santa Catarina, UDESC-Oeste, Chapecó 89815-630, SC, Brazil
| |
Collapse
|
7
|
Ma J, Wang PY, Zhuang J, Son AY, Karius AK, Syed AM, Nishi M, Wu Z, Mori MP, Kim YC, Hwang PM. CHCHD4-TRIAP1 regulation of innate immune signaling mediates skeletal muscle adaptation to exercise. Cell Rep 2024; 43:113626. [PMID: 38157298 PMCID: PMC10851177 DOI: 10.1016/j.celrep.2023.113626] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 10/20/2023] [Accepted: 12/11/2023] [Indexed: 01/03/2024] Open
Abstract
Exercise training can stimulate the formation of fatty-acid-oxidizing slow-twitch skeletal muscle fibers, which are inversely correlated with obesity, but the molecular mechanism underlying this transformation requires further elucidation. Here, we report that the downregulation of the mitochondrial disulfide relay carrier CHCHD4 by exercise training decreases the import of TP53-regulated inhibitor of apoptosis 1 (TRIAP1) into mitochondria, which can reduce cardiolipin levels and promote VDAC oligomerization in skeletal muscle. VDAC oligomerization, known to facilitate mtDNA release, can activate cGAS-STING/NFKB innate immune signaling and downregulate MyoD in skeletal muscle, thereby promoting the formation of oxidative slow-twitch fibers. In mice, CHCHD4 haploinsufficiency is sufficient to activate this pathway, leading to increased oxidative muscle fibers and decreased fat accumulation with aging. The identification of a specific mediator regulating muscle fiber transformation provides an opportunity to understand further the molecular underpinnings of complex metabolic conditions such as obesity and could have therapeutic implications.
Collapse
Affiliation(s)
- Jin Ma
- Cardiovascular Branch, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, MD 20892, USA
| | - Ping-Yuan Wang
- Cardiovascular Branch, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, MD 20892, USA
| | - Jie Zhuang
- Cardiovascular Branch, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, MD 20892, USA; School of Medicine, Nankai University, Tianjin 300071, China
| | - Annie Y Son
- Cardiovascular Branch, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, MD 20892, USA
| | - Alexander K Karius
- Cardiovascular Branch, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, MD 20892, USA
| | - Abu Mohammad Syed
- Cardiovascular Branch, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, MD 20892, USA
| | - Masahiro Nishi
- Cardiovascular Branch, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, MD 20892, USA
| | - Zhichao Wu
- Laboratory of Pathology, National Cancer Institute (NCI), NIH, Bethesda, MD 20892, USA
| | - Mateus P Mori
- Cardiovascular Branch, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, MD 20892, USA
| | - Young-Chae Kim
- Cardiovascular Branch, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, MD 20892, USA
| | - Paul M Hwang
- Cardiovascular Branch, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
8
|
Mackiewicz J, Lisek M, Boczek T. Targeting CaN/NFAT in Alzheimer's brain degeneration. Front Immunol 2023; 14:1281882. [PMID: 38077352 PMCID: PMC10701682 DOI: 10.3389/fimmu.2023.1281882] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/06/2023] [Indexed: 12/18/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by a progressive loss of cognitive functions. While the exact causes of this debilitating disorder remain elusive, numerous investigations have characterized its two core pathologies: the presence of β-amyloid plaques and tau tangles. Additionally, multiple studies of postmortem brain tissue, as well as results from AD preclinical models, have consistently demonstrated the presence of a sustained inflammatory response. As the persistent immune response is associated with neurodegeneration, it became clear that it may also exacerbate other AD pathologies, providing a link between the initial deposition of β-amyloid plaques and the later development of neurofibrillary tangles. Initially discovered in T cells, the nuclear factor of activated T-cells (NFAT) is one of the main transcription factors driving the expression of inflammatory genes and thus regulating immune responses. NFAT-dependent production of inflammatory mediators is controlled by Ca2+-dependent protein phosphatase calcineurin (CaN), which dephosphorylates NFAT and promotes its transcriptional activity. A substantial body of evidence has demonstrated that aberrant CaN/NFAT signaling is linked to several pathologies observed in AD, including neuronal apoptosis, synaptic deficits, and glia activation. In view of this, the role of NFAT isoforms in AD has been linked to disease progression at different stages, some of which are paralleled to diminished cognitive status. The use of classical inhibitors of CaN/NFAT signaling, such as tacrolimus or cyclosporine, or adeno-associated viruses to specifically inhibit astrocytic NFAT activation, has alleviated some symptoms of AD by diminishing β-amyloid neurotoxicity and neuroinflammation. In this article, we discuss the recent findings related to the contribution of CaN/NFAT signaling to the progression of AD and highlight the possible benefits of targeting this pathway in AD treatment.
Collapse
Affiliation(s)
| | | | - Tomasz Boczek
- Department of Molecular Neurochemistry, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
9
|
Danowska M, Strączkowski M. The Ca2+/Calmodulin-dependent Calcineurin/NFAT Signaling Pathway in the Pathogenesis of Insulin Resistance in Skeletal Muscle. Exp Clin Endocrinol Diabetes 2023; 131:589-594. [PMID: 37875146 DOI: 10.1055/a-2174-7958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2023]
Abstract
Skeletal muscle is the tissue directly involved in insulin-stimulated glucose uptake. Glucose is the primary energy substrate for contracting muscles, and proper metabolism of glucose is essential for health. Contractile activity and the associated Ca2+signaling regulate functional capacity and muscle mass. A high concentration of Ca2+and the presence of calmodulin (CaM) leads to the activation of calcineurin (CaN), a protein with serine-threonine phosphatase activity. The signaling pathway linked with CaN and transcription factors like the nuclear factor of activated T cells (NFAT) is essential for skeletal muscle development and reprogramming of fast-twitch to slow-twitch fibers. CaN activation may promote metabolic adaptations in muscle cells, resulting in better insulin-stimulated glucose transport. The molecular mechanisms underlying the altered insulin response remain unclear. The role of the CaN/NFAT pathway in regulating skeletal muscle hypertrophy is better described than its involvement in the pathogenesis of insulin resistance. Thus, there are opportunities for future research in that field. This review presents the role of CaN/NFAT signaling and suggests the relationship with insulin-resistant muscles.
Collapse
Affiliation(s)
- Magdalena Danowska
- Department of Prophylaxis of Metabolic Diseases, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Marek Strączkowski
- Department of Prophylaxis of Metabolic Diseases, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| |
Collapse
|
10
|
Protasi F, Girolami B, Roccabianca S, Rossi D. Store-operated calcium entry: From physiology to tubular aggregate myopathy. Curr Opin Pharmacol 2023; 68:102347. [PMID: 36608411 DOI: 10.1016/j.coph.2022.102347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 11/30/2022] [Accepted: 12/04/2022] [Indexed: 01/06/2023]
Abstract
Store-Operated Ca2+ entry (SOCE) is recognized as a key mechanism in muscle physiology necessary to refill intracellular Ca2+ stores during sustained muscle activity. For many years the cell structures expected to mediate SOCE in skeletal muscle fibres remained unknown. Recently, the identification of Ca2+ Entry Units (CEUs) in exercised muscle fibres opened new insights into the role of extracellular Ca2+ in muscle contraction and, more generally, in intracellular Ca2+ homeostasis. Accordingly, intracellular Ca2+ unbalance due to alterations in SOCE strictly correlates with muscle disfunction and disease. Mutations in proteins involved in SOCE (STIM1, ORAI1, and CASQ1) have been linked to tubular aggregate myopathy (TAM), a disease that causes muscle weakness and myalgia and is characterized by a typical accumulation of highly ordered and packed membrane tubules originated from the sarcoplasmic reticulum (SR). Achieving a full understanding of the molecular pathways activated by alterations in Ca2+ entry mechanisms is a necessary step to design effective therapies for human SOCE-related disorders.
Collapse
Affiliation(s)
- Feliciano Protasi
- CAST, Center for Advanced Studies and Technology; University G. d'Annunzio of Chieti-Pescara, I-66100, Italy; DMSI, Department of Medicine and Aging Sciences; University G. d'Annunzio of Chieti-Pescara, I-66100, Italy
| | - Barbara Girolami
- CAST, Center for Advanced Studies and Technology; University G. d'Annunzio of Chieti-Pescara, I-66100, Italy; DMSI, Department of Medicine and Aging Sciences; University G. d'Annunzio of Chieti-Pescara, I-66100, Italy
| | - Sara Roccabianca
- DMMS, Department of Molecular and Developmental Medicine; University of Siena, I-53100, Siena Italy
| | - Daniela Rossi
- DMMS, Department of Molecular and Developmental Medicine; University of Siena, I-53100, Siena Italy.
| |
Collapse
|
11
|
Kemi OJ. Exercise and Calcium in the Heart. CURRENT OPINION IN PHYSIOLOGY 2023. [DOI: 10.1016/j.cophys.2023.100644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
12
|
Zha W, Sun Y, Gong W, Li L, Kim W, Li H. Ginseng and ginsenosides: Therapeutic potential for sarcopenia. Biomed Pharmacother 2022; 156:113876. [DOI: 10.1016/j.biopha.2022.113876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/05/2022] [Accepted: 10/13/2022] [Indexed: 11/02/2022] Open
|
13
|
Kanakachari M, Ashwini R, Chatterjee RN, Bhattacharya TK. Embryonic transcriptome unravels mechanisms and pathways underlying embryonic development with respect to muscle growth, egg production, and plumage formation in native and broiler chickens. Front Genet 2022; 13:990849. [PMID: 36313432 PMCID: PMC9616467 DOI: 10.3389/fgene.2022.990849] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 09/12/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Muscle development, egg production, and plumage colors are different between native and broiler chickens. The study was designed to investigate why improved Aseel (PD4) is colorful, stronger, and grew slowly compared with the control broiler (CB). Methods: A microarray was conducted using the 7th-day embryo (7EB) and 18th-day thigh muscle (18TM) of improved Aseel and broiler, respectively. Also, we have selected 24 Gallus gallus candidate reference genes from NCBI, and total RNA was isolated from the broiler, improved Aseel embryo tissues, and their expression profiles were studied by real-time quantitative PCR (qPCR). Furthermore, microarray data were validated with qPCR using improved Aseel and broiler embryo tissues. Results: In the differential transcripts screening, all the transcripts obtained by microarray of slow and fast growth groups were screened by fold change ≥ 1 and false discovery rate (FDR) ≤ 0.05. In total, 8,069 transcripts were differentially expressed between the 7EB and 18TM of PD4 compared to the CB. A further analysis showed that a high number of transcripts are differentially regulated in the 7EB of PD4 (6,896) and fewer transcripts are differentially regulated (1,173) in the 18TM of PD4 compared to the CB. On the 7th- and 18th-day PD4 embryos, 3,890, 3,006, 745, and 428 transcripts were up- and downregulated, respectively. The commonly up- and downregulated transcripts are 91 and 44 between the 7th- and 18th-day of embryos. In addition, the best housekeeping gene was identified. Furthermore, we validated the differentially expressed genes (DEGs) related to muscle growth, myostatin signaling and development, and fatty acid metabolism genes in PD4 and CB embryo tissues by qPCR, and the results correlated with microarray expression data. Conclusion: Our study identified DEGs that regulate the myostatin signaling and differentiation pathway; glycolysis and gluconeogenesis; fatty acid metabolism; Jak-STAT, mTOR, and TGF-β signaling pathways; tryptophan metabolism; and PI3K-Akt signaling pathways in PD4. The results revealed that the gene expression architecture is present in the improved Aseel exhibiting embryo growth that will help improve muscle development, differentiation, egg production, protein synthesis, and plumage formation in PD4 native chickens. Our findings may be used as a model for improving the growth in Aseel as well as optimizing the growth in the broiler.
Collapse
Affiliation(s)
- M. Kanakachari
- ICAR-Directorate of Poultry Research, Hyderabad, India
- EVA.4 Unit, Faculty of Forestry and Wood Sciences, Czech University of Life Sciences Prague, Prague, Czechia
| | - R. Ashwini
- ICAR-Directorate of Poultry Research, Hyderabad, India
| | | | - T. K. Bhattacharya
- ICAR-Directorate of Poultry Research, Hyderabad, India
- *Correspondence: T. K. Bhattacharya,
| |
Collapse
|
14
|
Yang Y, Wu Y, Ji M, Rong X, Zhang Y, Yang S, Lu C, Cai C, Gao P, Guo X, Li B, Cao G. The long non-coding RNA lncMYOZ2 mediates an AHCY/MYOZ2 axis to promote adipogenic differentiation in porcine preadipocytes. BMC Genomics 2022; 23:700. [PMID: 36221052 PMCID: PMC9552422 DOI: 10.1186/s12864-022-08923-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 09/29/2022] [Indexed: 11/10/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) play a vital role in regulating adipogenesis. However, the associated regulatory mechanisms have yet to be described in detail in pig. In this study, we demonstrate a critical role for lncMYOZ2 in adipogenesis from porcine preadipocytes. Specifically, lncMYOZ2 was more abundant in the adipose tissue of Mashen (fat-type) pigs than for Large White (lean-type) pigs, and knockdown of this lncRNA significantly inhibited the differentiation of porcine preadipocytes into adipocytes. Mechanistically, we used RNA pull-down and RIP assays to establish that lncMYOZ2 interacts with adenosylhomocysteinase (AHCY). Moreover, lncMYOZ2 knockdown increased promoter methylation of the target gene MYOZ2 and lowered its expression. Finally, we describe a positive regulatory role for MYOZ2 in adipogenesis. Collectively, these findings establish lncMYOZ2 as an important epigenetic regulator of adipogenesis via the aforementioned AHCY/MYOZ2 pathway, and provide insights into the role of lncRNAs in porcine adipose development.
Collapse
Affiliation(s)
- Yang Yang
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Yiqi Wu
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Mengting Ji
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Xiaoyin Rong
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Yanwei Zhang
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Shuai Yang
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Chang Lu
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Chunbo Cai
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Pengfei Gao
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Xiaohong Guo
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Bugao Li
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Guoqing Cao
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China.
| |
Collapse
|
15
|
Zhen H, Zheng M, Geng H, Song Q, Gao L, Yuan Z, Deng H, Pang Q, Zhao B. The feedback loop between calcineurin, calmodulin-dependent protein kinase II, and nuclear factor of activated T-cells regulates the number of GABAergic neurons during planarian head regeneration. Front Mol Neurosci 2022; 15:988803. [PMID: 36172263 PMCID: PMC9510629 DOI: 10.3389/fnmol.2022.988803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/24/2022] [Indexed: 11/30/2022] Open
Abstract
Disturbances in the excitatory/inhibitory balance of brain neural circuits are the main source of encephalopathy during neurodevelopment. Changes in the function of neural circuits can lead to depolarization or repeat rhythmic firing of neurons in a manner similar to epilepsy. GABAergic neurons are inhibitory neurons found in all the main domains of the CNS. Previous studies suggested that DjCamkII and DjCaln play a crucial role in the regulation of GABAergic neurons during planarian regeneration. However, the mechanisms behind the regeneration of GABAergic neurons have not been fully explained. Herein, we demonstrated that DjCamkII and DjCaln were mutual negative regulation during planarian head regeneration. DjNFAT exerted feedback positive regulation on both DjCaln and DjCamkII. Whole-mount in situ hybridization (WISH) and fluorescence in situ hybridization (FISH) revealed that DjNFAT was predominantly expressed in the pharynx and parenchymal cells in intact planarian. Interestingly, during planarian head regeneration, DjNFAT was predominantly located in the newborn brain. Down-regulation of DjNFAT led to regeneration defects in the brain including regenerative brain became small and the lateral nerves cannot be regenerated completely, and a decreasein the number of GABAergic neurons during planarian head regeneration. These findings suggest that the feedback loop between DjCaln, DjCamkII, and DjNFAT is crucial for the formation of GABAergic neurons during planarian head regeneration.
Collapse
Affiliation(s)
- Hui Zhen
- Laboratory of Developmental and Evolutionary Biology, Shandong University of Technology, Nantong, China
| | - Mingyue Zheng
- Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Huazhi Geng
- Zibo Maternal and Child Health Hospital, Zibo, China
| | - Qian Song
- Laboratory of Developmental and Evolutionary Biology, Shandong University of Technology, Nantong, China
| | - Lili Gao
- Laboratory of Developmental and Evolutionary Biology, Shandong University of Technology, Nantong, China
| | - Zuoqing Yuan
- Laboratory of Developmental and Evolutionary Biology, Shandong University of Technology, Nantong, China
| | - Hongkuan Deng
- Laboratory of Developmental and Evolutionary Biology, Shandong University of Technology, Nantong, China
| | - Qiuxiang Pang
- Laboratory of Developmental and Evolutionary Biology, Shandong University of Technology, Nantong, China
| | - Bosheng Zhao
- Laboratory of Developmental and Evolutionary Biology, Shandong University of Technology, Nantong, China
- *Correspondence: Bosheng Zhao,
| |
Collapse
|
16
|
Chen L, Song M, Yao C. Calcineurin in development and disease. Genes Dis 2022; 9:915-927. [PMID: 35685477 PMCID: PMC9170610 DOI: 10.1016/j.gendis.2021.03.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/27/2021] [Accepted: 03/05/2021] [Indexed: 12/26/2022] Open
Abstract
Calcineurin (CaN) is a unique calcium (Ca2+) and calmodulin (CaM)-dependent serine/threonine phosphatase that becomes activated in the presence of increased intracellular Ca2+ level. CaN then functions to dephosphorylate target substrates including various transcription factors, receptors, and channels. Once activated, the CaN signaling pathway participates in the development of multiple organs as well as the onset and progression of various diseases via regulation of different cellular processes. Here, we review current literature regarding the structural and functional properties of CaN, highlighting its crucial role in the development and pathogenesis of immune system disorders, neurodegenerative diseases, kidney disease, cardiomyopathy and cancer.
Collapse
Affiliation(s)
- Lei Chen
- Department of Blood Transfusion, First Affiliated Hospital, The Third Military Medical University (Army Medical University), Chongqing 400038, PR China
| | - Min Song
- Department of Blood Transfusion, First Affiliated Hospital, The Third Military Medical University (Army Medical University), Chongqing 400038, PR China
| | - Chunyan Yao
- Department of Blood Transfusion, First Affiliated Hospital, The Third Military Medical University (Army Medical University), Chongqing 400038, PR China
| |
Collapse
|
17
|
The optimized core peptide derived from CABIN1 efficiently inhibits calcineurin-mediated T-cell activation. Exp Mol Med 2022; 54:613-625. [PMID: 35550603 PMCID: PMC9166766 DOI: 10.1038/s12276-022-00772-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 01/11/2022] [Accepted: 02/15/2022] [Indexed: 11/08/2022] Open
Abstract
The C-terminal fragment of CABIN1 interacts with calcineurin and represses the transcriptional activity of the nuclear factor of activated T cells (NFAT). However, the specific sequences and mechanisms through which it binds to calcineurin are unclear. This study determined that decameric peptide (CABIN1 residues 2146–2155) is minimally required for binding to calcineurin. This peptide contains a unique “PPTP” C-terminal sequence and a “PxIxIT” N-terminal motif. Furthermore, p38MAPK phosphorylated the threonine residue of the “PPTP” sequence under physiological conditions, dramatically enhancing the peptide’s binding affinity to calcineurin. Therefore, the CABIN1 peptide inhibited the calcineurin-NFAT pathway and the activation of T cells more efficiently than the VIVIT peptide without affecting calcineurin’s phosphatase activity. The CABIN1 peptide could thus be a more potent calcineurin inhibitor and provide therapeutic opportunities for various diseases caused by the calcineurin-NFAT pathway. A peptide with therapeutic potential binds strongly to the cellular enzyme calcineurin and may prove valuable in anti-cancer and autoimmune disease treatments. Many cancers and autoimmune diseases are linked with overactivation of a key calcineurin-related pathway which is heavily involved in T cell activation. This pathway has long been a therapeutic target, but existing drugs show problems with stability and delivery, and can cause serious side effects. One known inhibitor of calcineurin is the protein CABIN1, but precisely how well it binds and how useful it may be is unclear. Now, Hong-Duk Youn at Seoul National University College of Medicine, South Korea, and co-workers have identified how one specific peptide from CABIN1 binds strongly to calcineurin. The CABIN1 peptide was stable and displayed greater efficiency at inhibiting calcineurin than another recently identified peptide candidate.
Collapse
|
18
|
Cao H, Zhou Q, Liu C, Zhang Y, Xie M, Qiao W, Dong N. Substrate stiffness regulates differentiation of induced pluripotent stem cells into heart valve endothelial cells. Acta Biomater 2022; 143:115-126. [PMID: 35235867 DOI: 10.1016/j.actbio.2022.02.032] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 02/08/2022] [Accepted: 02/21/2022] [Indexed: 12/15/2022]
Abstract
Substrate stiffness has been indicated as a primary determinant for stem cell fate, being capable of influencing motility, proliferation, and differentiation. Although the effects of stiffness on cardiac differentiation of human-induced pluripotent stem cells (h-iPSCs) have been reported, whether stiffness of polydimethylsiloxane-based substrates could enhance differentiation of h-iPSCs toward heart valve endothelial cells lineage (VECs) or not remains unknown. Herein, we modulated the substrate stiffness to evaluate its effect on the differentiation of h-iPSCs into valve endothelial-like cells (h-iVECs) in vitro and determine the suitable stiffness. The results revealed that VECs-related genes (PECAM1, CDH5, NFATC1, etc.) were significantly increased in h-iVECs obtained from the three substrates compared with h-iPSCs. Gene expression levels and differentiation efficiency were higher in the medium group than in the stiff and soft groups. An increase in substrate stiffness to 2.8 GPa decreased the efficiency of h-iPSCs differentiation into h-iVECs and downregulated VECs specific genes. Through mRNA sequencing, we determined the key genetic markers involved in stiffness guiding the differentiation of cardiac progenitor cells into h-iVECs. Unsupervised hierarchical clustering showed that medium stiffness were more suitable for the differentiation of h-iPSCs into h-iVECs in vitro. Moreover, this process is regulated by the WNT/Calcineurin signaling pathway. Overall, this study demonstrates how stiffness can be used to enhance the h-iVECs differentiation of iPSCs and emphasizes the importance of using substrate stiffness to accomplish a more specific and mature differentiation of h-iVECs for future therapeutic and tissue engineering valve applications. STATEMENT OF SIGNIFICANCE: Several studies have examined the stiffness-induced cell fate from pluripotent stem cells during the stage of mesoderm cell differentiation. This is the first research that rigorously examines the effect of substrate stiffness on human valve endothelial-like cells differentiation from cardiac progenitor cells. We found that the medium stiffness can increase the differentiation efficiency of h-iVECs from 40% to about 60%, and this process was regulated by the WNT/CaN signaling pathway through the activation of WNT5a. Substrate stiffness not only increases the differentiation efficiency of h-iVECs, but also improves its cellular functions such as low-density lipoprotein uptake and NO release. This study emphasizes the importance of using substrate stiffness to accomplish a more specific and mature differentiation of h-iVECs.
Collapse
|
19
|
The role of long noncoding RNA Nron in atherosclerosis development and plaque stability. iScience 2022; 25:103978. [PMID: 35295812 PMCID: PMC8919297 DOI: 10.1016/j.isci.2022.103978] [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: 09/27/2021] [Revised: 12/31/2021] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
The major clinical consequences of atherosclerosis such as myocardial infarction or stroke are because of thrombotic events associated with acute rupture or erosion of an unstable plaque. Here, we identify an lncRNA Noncoding Repressor of NFAT (Nron) as a critical regulator of atherosclerotic plaque stability. Nron overexpression (OE) in vascular smooth muscle cells (VSMC) induces a highly characteristic architecture of more-vulnerable plaques, while Nron knockdown (KD) suppresses the development of atherosclerosis and favors plaque stability. Mechanistically, Nron specifically binds to and negatively regulates NFATc3, thus inhibiting the proliferation and promoting the apoptosis of VSMCs. Moreover, we also provide evidence that Nron increases the production and secretion of VEGFA from VSMCs, which functions as a paracrine factor to enhance intra-plaque angiogenesis. All of these effects contribute to plaque instability. Genetic or pharmacological inhibition of Nron may have potential for future therapy of atherosclerosis. Nron promotes atherosclerosis progression and contributes to plaque instability Nron negatively regulates NFATc3 activity and impairs VSMC function Nron increases VEGFA production and promotes intra-plaque angiogenesis
Collapse
|
20
|
Attwaters M, Hughes SM. Cellular and molecular pathways controlling muscle size in response to exercise. FEBS J 2022; 289:1428-1456. [PMID: 33755332 DOI: 10.1111/febs.15820] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 02/27/2021] [Accepted: 03/12/2021] [Indexed: 12/14/2022]
Abstract
From the discovery of ATP and motor proteins to synaptic neurotransmitters and growth factor control of cell differentiation, skeletal muscle has provided an extreme model system in which to understand aspects of tissue function. Muscle is one of the few tissues that can undergo both increase and decrease in size during everyday life. Muscle size depends on its contractile activity, but the precise cellular and molecular pathway(s) by which the activity stimulus influences muscle size and strength remain unclear. Four correlates of muscle contraction could, in theory, regulate muscle growth: nerve-derived signals, cytoplasmic calcium dynamics, the rate of ATP consumption and physical force. Here, we summarise the evidence for and against each stimulus and what is known or remains unclear concerning their molecular signal transduction pathways and cellular effects. Skeletal muscle can grow in three ways, by generation of new syncytial fibres, addition of nuclei from muscle stem cells to existing fibres or increase in cytoplasmic volume/nucleus. Evidence suggests the latter two processes contribute to exercise-induced growth. Fibre growth requires increase in sarcolemmal surface area and cytoplasmic volume at different rates. It has long been known that high-force exercise is a particularly effective growth stimulus, but how this stimulus is sensed and drives coordinated growth that is appropriately scaled across organelles remains a mystery.
Collapse
Affiliation(s)
- Michael Attwaters
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, UK
| | - Simon M Hughes
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, UK
| |
Collapse
|
21
|
Ritchie JA, Ng JQ, Kemi OJ. When one says yes and the other says no; does calcineurin participate in physiologic cardiac hypertrophy? ADVANCES IN PHYSIOLOGY EDUCATION 2022; 46:84-95. [PMID: 34762541 DOI: 10.1152/advan.00104.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 11/08/2021] [Indexed: 06/13/2023]
Abstract
Developing engaging activities that build skills for understanding and appreciating research is important for undergraduate and postgraduate science students. Comparing and contrasting opposing research studies does this, and more: it also appropriately for these cohorts challenges higher level cognitive processing. Here, we present and discuss one such scenario, that of calcineurin in the heart and its response to exercise training. This scenario is further accentuated by the existence of only two studies. The background is that regular aerobic endurance exercise training stimulates the heart to physiologically adapt to chronically increase its ability to produce a greater cardiac output to meet the increased demand for oxygenated blood in working muscles, and this happens by two main mechanisms: 1) increased cardiac contractile function and 2) physiologic hypertrophy. The major underlying mechanisms have been delineated over the last decades, but one aspect has not been resolved: the potential role of calcineurin in modulating physiologic hypertrophy. This is partly because the existing research has provided opposing and contrasting findings, one line showing that exercise training does activate cardiac calcineurin in conjunction with myocardial hypertrophy, but another line showing that exercise training does not activate cardiac calcineurin even if myocardial hypertrophy is blatantly occurring. Here, we review and present the current evidence in the field and discuss reasons for this controversy. We present real-life examples from physiology research and discuss how this may enhance student engagement and participation, widen the scope of learning, and thereby also further facilitate higher level cognitive processing.
Collapse
Affiliation(s)
- Jonathan A Ritchie
- School of Medicine, Dentistry and Nursing, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Jun Q Ng
- School of Life Sciences, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Ole J Kemi
- School of Life Sciences, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
22
|
Taylor DF, Bishop DJ. Transcription Factor Movement and Exercise-Induced Mitochondrial Biogenesis in Human Skeletal Muscle: Current Knowledge and Future Perspectives. Int J Mol Sci 2022; 23:1517. [PMID: 35163441 PMCID: PMC8836245 DOI: 10.3390/ijms23031517] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/19/2022] [Accepted: 01/21/2022] [Indexed: 02/01/2023] Open
Abstract
In response to exercise, the oxidative capacity of mitochondria within skeletal muscle increases through the coordinated expression of mitochondrial proteins in a process termed mitochondrial biogenesis. Controlling the expression of mitochondrial proteins are transcription factors-a group of proteins that regulate messenger RNA transcription from DNA in the nucleus and mitochondria. To fulfil other functions or to limit gene expression, transcription factors are often localised away from DNA to different subcellular compartments and undergo rapid movement or accumulation only when required. Although many transcription factors involved in exercise-induced mitochondrial biogenesis have been identified, numerous conflicting findings and gaps exist within our knowledge of their subcellular movement. This review aims to summarise and provide a critical analysis of the published literature regarding the exercise-induced movement of transcription factors involved in mitochondria biogenesis in skeletal muscle.
Collapse
Affiliation(s)
| | - David J. Bishop
- Institute for Health and Sport (iHeS), Footscray Park, Victoria University, Melbourne 8001, Australia;
| |
Collapse
|
23
|
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: 5.0] [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
|
24
|
Nolze A, Köhler C, Ruhs S, Quarch K, Strätz N, Gekle M, Grossmann C. Calcineurin (PPP3CB) regulates angiotensin II-dependent vascular remodelling by potentiating EGFR signalling in mice. Acta Physiol (Oxf) 2021; 233:e13715. [PMID: 34228904 DOI: 10.1111/apha.13715] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 06/08/2021] [Accepted: 07/03/2021] [Indexed: 12/14/2022]
Abstract
AIM This study investigates the role of calcineurin for angiotensin II (AngII)-induced vascular remodelling with the help of a mouse model lacking the catalytic beta subunit of calcineurin (PPP3CB KO). METHODS Wildtype (WT) and PPP3CB KO mice were treated for 4 weeks with AngII followed by assessment of blood pressure, histological evaluation of aortas and mRNA analysis of aortic genes PPP3CB-dependently regulated by AngII. Primary murine vascular smooth muscle cells (VSMCs) were used for qPCR, ELISA and Western Blot experiments as well as wound healing and cell proliferation assays. RESULTS Upon AngII treatment, PPP3CB KO mice showed less aortic media thickening, lumen dilation and systolic blood pressure compared to WT mice. Next-generation sequencing data of aortic tissue indicated an increase in extracellular matrix components (EMCs), cell migration and cell proliferation. A PPP3CB-dependent increase in EMC was confirmed by qPCR in aorta and VSMCs. PPP3CB-dependent stimulation of VSMC migration could be verified by wound healing assays but markers of enhanced cell proliferation were only detectable in aortic tissue of WT mice but not in isolated WT or KO VSMCs. We could demonstrate in VSMCs with pharmacological inhibitors that PPP3CB leads to enhanced heparin-binding EGF-like growth factor (HB-EGF) secretion, epidermal growth factor receptor (EGFR) activation and consecutive stimulation of transforming growth factor β(TGFβ) and connective tissue growth factor (CTGF) signalling that enhances collagen expression. CONCLUSION AngII-induced vascular remodelling involves PPP3CB, which leads to enhanced EMC production, VSMC migration and sustained increase in systolic blood pressure via HBEGF/EGFR-TGFβ-CTGF signalling.
Collapse
Affiliation(s)
- Alexander Nolze
- Julius‐Bernstein‐Institute of PhysiologyMartin Luther University Halle‐Wittenberg Halle Germany
| | - Conny Köhler
- Julius‐Bernstein‐Institute of PhysiologyMartin Luther University Halle‐Wittenberg Halle Germany
| | - Stefanie Ruhs
- Julius‐Bernstein‐Institute of PhysiologyMartin Luther University Halle‐Wittenberg Halle Germany
| | - Katja Quarch
- Julius‐Bernstein‐Institute of PhysiologyMartin Luther University Halle‐Wittenberg Halle Germany
| | - Nicole Strätz
- Julius‐Bernstein‐Institute of PhysiologyMartin Luther University Halle‐Wittenberg Halle Germany
| | - Michael Gekle
- Julius‐Bernstein‐Institute of PhysiologyMartin Luther University Halle‐Wittenberg Halle Germany
| | - Claudia Grossmann
- Julius‐Bernstein‐Institute of PhysiologyMartin Luther University Halle‐Wittenberg Halle Germany
| |
Collapse
|
25
|
Ulengin-Talkish I, Parson MAH, Jenkins ML, Roy J, Shih AZL, St-Denis N, Gulyas G, Balla T, Gingras AC, Várnai P, Conibear E, Burke JE, Cyert MS. Palmitoylation targets the calcineurin phosphatase to the phosphatidylinositol 4-kinase complex at the plasma membrane. Nat Commun 2021; 12:6064. [PMID: 34663815 PMCID: PMC8523714 DOI: 10.1038/s41467-021-26326-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 09/29/2021] [Indexed: 11/25/2022] Open
Abstract
Calcineurin, the conserved protein phosphatase and target of immunosuppressants, is a critical mediator of Ca2+ signaling. Here, to discover calcineurin-regulated processes we examined an understudied isoform, CNAβ1. We show that unlike canonical cytosolic calcineurin, CNAβ1 localizes to the plasma membrane and Golgi due to palmitoylation of its divergent C-terminal tail, which is reversed by the ABHD17A depalmitoylase. Palmitoylation targets CNAβ1 to a distinct set of membrane-associated interactors including the phosphatidylinositol 4-kinase (PI4KA) complex containing EFR3B, PI4KA, TTC7B and FAM126A. Hydrogen-deuterium exchange reveals multiple calcineurin-PI4KA complex contacts, including a calcineurin-binding peptide motif in the disordered tail of FAM126A, which we establish as a calcineurin substrate. Calcineurin inhibitors decrease PI4P production during Gq-coupled GPCR signaling, suggesting that calcineurin dephosphorylates and promotes PI4KA complex activity. In sum, this work discovers a calcineurin-regulated signaling pathway which highlights the PI4KA complex as a regulatory target and reveals that dynamic palmitoylation confers unique localization, substrate specificity and regulation to CNAβ1.
Collapse
Affiliation(s)
| | - Matthew A H Parson
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | - Meredith L Jenkins
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | - Jagoree Roy
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Alexis Z L Shih
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada
- Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Nicole St-Denis
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, University of Toronto, Toronto, Canada
- High-Fidelity Science Communications, Summerside, PE, Canada
| | - Gergo Gulyas
- Section on Molecular Signal Transduction, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Tamas Balla
- Section on Molecular Signal Transduction, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, University of Toronto, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Péter Várnai
- Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Elizabeth Conibear
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - John E Burke
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
- Department of Biochemistry, The University of British Columbia, Vancouver, BC, Canada
| | - Martha S Cyert
- Department of Biology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
26
|
Fukui H, Chow RWY, Xie J, Foo YY, Yap CH, Minc N, Mochizuki N, Vermot J. Bioelectric signaling and the control of cardiac cell identity in response to mechanical forces. Science 2021; 374:351-354. [PMID: 34648325 DOI: 10.1126/science.abc6229] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Hajime Fukui
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Centre National de la Recherche Scientifique UMR7104, Institut National de la Santé et de la Recherche Médicale U1258 and Université de Strasbourg, Illkirch, France.,Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Renee Wei-Yan Chow
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Centre National de la Recherche Scientifique UMR7104, Institut National de la Santé et de la Recherche Médicale U1258 and Université de Strasbourg, Illkirch, France
| | - Jing Xie
- Université de Paris, Centre National de la Recherche Scientifique UMR7592, Institut Jacques Monod, Paris, France
| | - Yoke Yin Foo
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore
| | - Choon Hwai Yap
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore.,Department of Bioengineering, Imperial College London, London, UK
| | - Nicolas Minc
- Université de Paris, Centre National de la Recherche Scientifique UMR7592, Institut Jacques Monod, Paris, France
| | - Naoki Mochizuki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Julien Vermot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Centre National de la Recherche Scientifique UMR7104, Institut National de la Santé et de la Recherche Médicale U1258 and Université de Strasbourg, Illkirch, France.,Department of Bioengineering, Imperial College London, London, UK
| |
Collapse
|
27
|
Ni L, Lahiri SK, Nie J, Pan X, Abu-Taha I, Reynolds JO, Campbell HM, Wang H, Kamler M, Schmitz W, Müller FU, Li N, Wei X, Wang DW, Dobrev D, Wehrens XHT. Genetic inhibition of Nuclear Factor of Activated T-cell c2 (NFATc2) prevents atrial fibrillation in CREM transgenic mice. Cardiovasc Res 2021; 118:2805-2818. [PMID: 34648001 PMCID: PMC9586567 DOI: 10.1093/cvr/cvab325] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 10/11/2021] [Indexed: 11/14/2022] Open
Abstract
AIMS Abnormal intracellular calcium handling contributes to the progressive nature of atrial fibrillation (AF), the most common sustained cardiac arrhythmia. Evidence in mouse models suggests that activation of the nuclear factor of activated T-cell (NFAT) signaling pathway contributes to atrial remodeling. Our aim was to determine the role of NFATc2 in AF in humans and mouse models. METHODS AND RESULTS Expression levels of NFATc1-c4 isoforms were assessed by quantitative reverse transcription-polymerase chain reaction in right atrial appendages from patients with chronic AF. NFATc1 and NFATc2 mRNA levels were elevated in chronic AF (cAF) patients compared with those in sinus rhythm (SR). Western blotting revealed increased cytosolic and nuclear levels of NFATc2 in AF patients. Similar findings were obtained in CREM-IbΔC-X transgenic (CREM) mice, a model of progressive AF. Telemetry ECG recordings revealed age-dependent spontaneous AF in CREM mice, which was prevented by NFATc2 knockout in CREM: NFATc2-/- mice. Programmed electrical stimulation revealed that CREM: NFATc2-/- mice lacked an AF substrate. Morphometric analysis and histology revealed increased atrial weight and atrial fibrosis in CREM mice compared with WT controls, which was reversed in CREM: NFATc2-/- mice. Confocal microscopy showed an increased Ca2+ spark frequency despite a reduced sarcoplasmic reticulum (SR) Ca2+ load in CREM mice compared with controls, whereas these abnormalities were normalized in CREM: NFATc2-/- mice. Western blotting revealed that genetic inhibition of Ca2+/calmodulin-dependent protein kinase II-mediated phosphorylation of S2814 on RyR2 in CREM: RyR2-S2814A mice suppressed NFATc2 activation observed in CREM mice, suggesting that NFATc2 is activated by excessive SR Ca2+ leak via RyR2. Finally, chromatin immunoprecipitation sequencing from AF patients identified Ras And EF-Hand Domain-Containing Protein (RASEF) as a direct target of NFATc2 mediated transcription. CONCLUSION Our findings reveal activation of the NFAT signaling pathway in patients of Chinese and European descent. NFATc2 knockout prevents the progression of AF in the CREM mouse model. TRANSLATIONAL PERSPECTIVE Atrial fibrillation (AF) is a progressive disease characterized by electrical and structural remodeling which promotes atrial arrhythmias. This study provides evidence for increased 'nuclear factor of activated T-cell' (NFAT) signaling in patients with chronic AF. Studies in the CREM transgenic model of progressive AF revealed that the NFATc2 isoform mediates atrial remodeling associated with AF substrate development. Chromatin immunoprecipitation sequencing of atrial biopsies from AF patients identified 'Ras And EF-Hand Domain-Containing Protein' (RASEF) as a downstream target of NFATc2-mediated transcription, suggesting that targeting these factors might be beneficial for curtailing AF progression.
Collapse
Affiliation(s)
- Li Ni
- Division of Cardiology, Department of Internal Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Satadru K Lahiri
- Cardiovascular Research Institute.,Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Jiali Nie
- Division of Cardiology, Department of Internal Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Xiaolu Pan
- Cardiovascular Research Institute.,Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Issam Abu-Taha
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Julia O Reynolds
- Cardiovascular Research Institute.,Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Hannah M Campbell
- Cardiovascular Research Institute.,Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Haihao Wang
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Markus Kamler
- Cardiac Surgery II Essen-Huttrop, University Hospital, West German Heart Center, University of Essen, Germany
| | - Wilhelm Schmitz
- Institute of Pharmacology and Toxicology, University of Münster, Germany
| | | | - Na Li
- Cardiovascular Research Institute.,Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA.,Institute of Pharmacology and Toxicology, University of Münster, Germany
| | - Xiang Wei
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Xander H T Wehrens
- Cardiovascular Research Institute.,Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA.,Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, Houston, TX, 77030 USA.,Department of Medicine (Cardiology), Baylor College of Medicine, Houston, TX, 77030 USA.,Department of Pediatrics, Center for Space Medicine, Baylor College of Medicine, Houston, TX, 77030 USA
| |
Collapse
|
28
|
Kim HJ, Kim H, Choi Y, Lee JH, Kim D, Lee SK, Park KS. Cinnamomum verum-derived O-methoxycinnamaldehyde prevents lipopolysaccharide-induced depressive-like behavior in mice via NFAT mRNA stability in T lymphocytes. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 91:153703. [PMID: 34425473 DOI: 10.1016/j.phymed.2021.153703] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/30/2021] [Accepted: 08/06/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Depressive-like behaviors are related to inflammatory immune activation. Cinnamomum verum (CV) has anti-inflammatory effects, but the molecular mechanisms underlying the antidepressant effects after immunological activation still remain elusive. PURPOSE The aim of the present study was to investigate the effect of CV in improving depressive-like behavior and explore its underlying mechanism in T lymphocytes. METHODS Mice were randomly divided into Control, LPS, LPS plus fluoxetine, LPS plus CV, and LPS plus MCA groups. Behavior was evaluated using forced swimming test (FST) and tail suspension test (TST). The experimental group mice were exposed to LPS to induce depressive-like behavior. Cell viability was measured upon treating splenic T lymphocytes and Jurkat T cells with CV. Cytokine activity was measured using ELISA and RT-qPCR. The components of CV were analyzed by HPLC. NFAT expression was evaluated by western blotting, immunofluorescence, and luciferase assay. To verify the half-life of NFAT mRNA, Jurkat cells were treated with actinomycin D for 1.5, 3, and 4.5 h. RESULTS CV effectively prevents inflammation-induced depressive-like behaviors. CV dose-dependently decreased protein and mRNA levels of TNFα and IL-2. Inhibition of TNFα and IL-2 production involves an MCA-mediated decrease in NFAT mRNA level, rather than inhibition of nuclear translocation. This mechanism was independent of NFAT transcription inducer p38 MAPK; it can be attributed to the promotion of NFAT mRNA decay. CONCLUSION Overall, MCA might be an alternative or adjuvant to existing NFAT-targeting immunosuppressants for clinical prophylaxis or therapy in the context of inflammation-induced depressive disorder or other T-cell-associated inflammatory disorders.
Collapse
Affiliation(s)
- Hye Jin Kim
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea; College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyungjun Kim
- KM Science Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Yujin Choi
- KM Science Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Jun-Hwan Lee
- KM Science Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea; Korean Medicine Life Science, University of Science & Technology (UST), Campus of Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Donghwan Kim
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Sang Kook Lee
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea.
| | - Ki-Sun Park
- KM Science Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea.
| |
Collapse
|
29
|
Taneja A, Ravi V, Hong JY, Lin H, Sundaresan NR. Emerging roles of Sirtuin 2 in cardiovascular diseases. FASEB J 2021; 35:e21841. [PMID: 34582046 DOI: 10.1096/fj.202100490r] [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: 03/24/2021] [Revised: 07/07/2021] [Accepted: 07/23/2021] [Indexed: 11/11/2022]
Abstract
Sirtuins are a family of NAD+ -dependent deacetylases implicated in a wide variety of age-associated pathologies, including cardiovascular disorders. Among the seven mammalian sirtuins, SIRT2 modulates various cellular processes through the deacetylation or deacylation of their target proteins. Notably, the levels of SIRT2 in the heart decline with age and other pathological conditions, leading to cardiovascular dysfunction. In the present review, we discuss the emerging roles of SIRT2 in cardiovascular dysfunction and heart failure associated with factors like age, hypertension, oxidative stress, and diabetes. We also discuss the potential of using inhibitors to study the unexplored role of SIRT2 in the heart. While SIRT2 undoubtedly plays a crucial role in the cardiovascular system, its functions are only beginning to be understood, making it an attractive candidate for further research in the field.
Collapse
Affiliation(s)
- Arushi Taneja
- Department of Microbiology and Cell Biology, Cardiovascular and Muscle Research Laboratory, Indian Institute of Science, Bengaluru, India
| | - Venkatraman Ravi
- Department of Microbiology and Cell Biology, Cardiovascular and Muscle Research Laboratory, Indian Institute of Science, Bengaluru, India
| | - Jun Young Hong
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, USA
| | - Hening Lin
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, USA.,Howard Hughes Medical Institute, Cornell University, Ithaca, NY, USA
| | - Nagalingam Ravi Sundaresan
- Department of Microbiology and Cell Biology, Cardiovascular and Muscle Research Laboratory, Indian Institute of Science, Bengaluru, India
| |
Collapse
|
30
|
Du M, Yang L, Liu B, Yang L, Mao X, Liang M, Huang K. Inhibition of NFAT suppresses foam cell formation and the development of diet-induced atherosclerosis. FASEB J 2021; 35:e21951. [PMID: 34551141 DOI: 10.1096/fj.202100947r] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 09/02/2021] [Accepted: 09/09/2021] [Indexed: 11/11/2022]
Abstract
Deciphering the molecular and cellular processes involved in foam cell formation is critical for us to understand the pathogenesis of atherosclerosis. Nuclear factor of activated T cells (NFAT) is a transcription factor originally identified as a key player in the differentiation of T cells and maturation of immune system. Nowadays it has been brought into attention that NFAT also regulates multiple pathophysiological processes and targeted intervention in NFAT may be effective in the treatment of some cardiovascular diseases. However, whether NFAT is involved in foam cell formation remains elusive. NFAT in human monocyte-derived macrophage was activated by ox-LDL and translocated from the cytoplasm to the nucleus. NFAT then directly bound to peroxisome proliferator-activated receptor γ (PPARγ) in the nucleus and negatively regulated its transcriptional activity. NFATc2 knockdown or NFAT inhibitor 11R-VIVIT increased cholesterol efflux (by activating PPARγ-LXRα-ABCA1 cascade) and reduced the uptake of modified lipoprotein (in a PPARγ-independent way) in macrophage, thus prevented foam cell formation. Besides, 11R-VIVIT also exerted a protective role in the development of atherosclerosis in western diet-fed ApoE-/- mice. These results suggest NFAT inhibition as a potential therapeutic strategy in atherosclerosis.
Collapse
Affiliation(s)
- Meng Du
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Liyuan Cardiovascular Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liu Yang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bing Liu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liuye Yang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxiang Mao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Minglu Liang
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Huang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Liyuan Cardiovascular Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
31
|
Tanaka M, Ikeji T, Nakanishi R, Hirabayashi T, Ono K, Hirayama Y, Tategaki A, Kondo H, Ishihara A, Fujino H. Protective effects of Enterococcus faecium strain R30 supplementation on decreased muscle endurance under disuse in rats. Exp Physiol 2021; 106:1961-1970. [PMID: 34216158 DOI: 10.1113/ep089677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 07/01/2021] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the central question of this study? Does Enterococcus faecium strain R30 (R30), a new lactic acid bacterial strain for supplementation, attenuate shifts in the typology of whole muscle fibres from slow- to fast-twitch by altering the autonomic nervous system in atrophied skeletal muscles? What is the main finding and its importance? R30 supplementation may attenuate the shifts in the typology of whole muscle fibres from slow- to fast-twitch fibres by upregulating peroxisome proliferator-activated receptor-γ coactivator-1α and activating the calcineurin-nuclear factor of activated T-cells signalling pathway, thus ameliorating the decrease in muscle endurance associated with disuse. ABSTRACT Enterococcus faecium strain R30 (R30), a new lactic acid bacterial strain for supplementation, was hypothesized to attenuate shifts in the typology of whole muscle fibres from slow- to fast-twitch fibres in atrophied skeletal muscles. We further postulated that the prevention of slow-to-fast fibre shifts would suppress the decreased muscle endurance associated with atrophy. To evaluate the protective effects of R30, we analysed slow-to-fast fibre shifts and disuse-associated reduced muscle endurance. R30 was administered to rats with an acclimation period of 7 days before hindlimb unloading (HU) for 2 weeks. The composition ratio of the fibre type and the expression levels of peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α), calcineurin and nuclear factor of activated T-cells (NFAT) were measured. Muscle endurance was evaluated at the end of the 2-week HU period in an in situ environment. R30 supplementation suppressed the slow-to-fast fibre switch and decreased the HU-induced expression of PGC-1α proteins and the deactivation of the calcineurin-NFAT pathway. Furthermore, R30 prevented a decrease in HU-associated muscle endurance in calf muscles. These results indicate that R30 supplementation may attenuate the shifts in the typology of whole muscle fibres from slow- to fast-twitch fibres via the upregulation of PGC-1α and the activation of the calcineurin-NFAT signalling pathway, thereby ameliorating the decrease in muscle endurance associated with disuse.
Collapse
Affiliation(s)
- Minoru Tanaka
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan.,Department of Rehabilitation Science, Osaka Health Science University, Osaka, Japan
| | - Takuya Ikeji
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Ryosuke Nakanishi
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan.,Faculty of Rehabilitation, Department of Physical Therapy, Kobe international University, Kobe, Japan
| | - Takumi Hirabayashi
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Kohei Ono
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Yusuke Hirayama
- Biotechnology Research Laboratories, Kaneka Corporation, Takasago, Japan
| | - Airo Tategaki
- Biotechnology Research Laboratories, Kaneka Corporation, Takasago, Japan
| | - Hiroyo Kondo
- Department of Food Science and Nutrition, Nagoya Women's University, Nagoya, Japan
| | - Akihiko Ishihara
- Graduate School of Human and Environmental Studies, Kyoto University, Kyoto, Japan
| | - Hidemi Fujino
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| |
Collapse
|
32
|
Zhang X, Li G, Deng Q, Xu Z, Cen J, Xu J. Vomifoliol isolated from mangrove plant Ceriops tagal inhibits the NFAT signaling pathway with CN as the target enzyme in vitro. Bioorg Med Chem Lett 2021; 48:128235. [PMID: 34216746 DOI: 10.1016/j.bmcl.2021.128235] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 06/17/2021] [Accepted: 06/26/2021] [Indexed: 11/26/2022]
Abstract
Vomifoliol, a natural sesquiterpene compound, is a secondary metabolite isolated from the mangrove plant Ceriops tagal. The present study aimed to determine the immunosuppressive effects and underlying mechanisms of vomifoliol on Jurkat cells in vitro. The results show that vomifoliol significantly inhibited calcineurin (CN) at concentrations resulting in relatively low cytotoxicity. Moreover, vomifoliol was found to exert an inhibitory effect on phorbol 12-myristate 13-acetate (PMA)/ ionomycin (Io) -induced Jurkat cells and the dephosphorylation of NFAT1. In addition, it reduced the expression of IL-2. Based on these results, we concluded that vomifoliol may inhibit the immune response of Jurkat cells, and vomifoliol may use CN as the target enzyme to inhibit NFAT signaling pathway. Therefore, vomifoliol may be promising as a low-toxic natural immunosuppressant.
Collapse
Affiliation(s)
- Xuexia Zhang
- Key Laboratory of Advanced Materials of Tropical Island Resources of Ministry of Education, School of Chemical Engineering and Technology, Hainan University, Haikou 570228, PR China
| | - Gang Li
- Key Laboratory of Advanced Materials of Tropical Island Resources of Ministry of Education, School of Chemical Engineering and Technology, Hainan University, Haikou 570228, PR China
| | - Qin Deng
- College of Horticulture and Landscape, Hainan University, Haikou 570228, PR China
| | - Zhiyong Xu
- School of Life and Pharmaceutical Sciences, Hainan University, Haikou 570228, PR China
| | - Juren Cen
- School of Life and Pharmaceutical Sciences, Hainan University, Haikou 570228, PR China.
| | - Jing Xu
- Key Laboratory of Advanced Materials of Tropical Island Resources of Ministry of Education, School of Chemical Engineering and Technology, Hainan University, Haikou 570228, PR China.
| |
Collapse
|
33
|
Basse AL, Agerholm M, Farup J, Dalbram E, Nielsen J, Ørtenblad N, Altıntaş A, Ehrlich AM, Krag T, Bruzzone S, Dall M, de Guia RM, Jensen JB, Møller AB, Karlsen A, Kjær M, Barrès R, Vissing J, Larsen S, Jessen N, Treebak JT. Nampt controls skeletal muscle development by maintaining Ca 2+ homeostasis and mitochondrial integrity. Mol Metab 2021; 53:101271. [PMID: 34119711 PMCID: PMC8259345 DOI: 10.1016/j.molmet.2021.101271] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/31/2021] [Accepted: 06/09/2021] [Indexed: 12/12/2022] Open
Abstract
Objective NAD+ is a co-factor and substrate for enzymes maintaining energy homeostasis. Nicotinamide phosphoribosyltransferase (NAMPT) controls NAD+ synthesis, and in skeletal muscle, NAD+ is essential for muscle integrity. However, the underlying molecular mechanisms by which NAD+ synthesis affects muscle health remain poorly understood. Thus, the objective of the current study was to delineate the role of NAMPT-mediated NAD+ biosynthesis in skeletal muscle development and function. Methods To determine the role of Nampt in muscle development and function, we generated skeletal muscle-specific Nampt KO (SMNKO) mice. We performed a comprehensive phenotypic characterization of the SMNKO mice, including metabolic measurements, histological examinations, and RNA sequencing analyses of skeletal muscle from SMNKO mice and WT littermates. Results SMNKO mice were smaller, with phenotypic changes in skeletal muscle, including reduced fiber area and increased number of centralized nuclei. The majority of SMNKO mice died prematurely. Transcriptomic analysis identified that the gene encoding the mitochondrial permeability transition pore (mPTP) regulator Cyclophilin D (Ppif) was upregulated in skeletal muscle of SMNKO mice from 2 weeks of age, with associated increased sensitivity of mitochondria to the Ca2+-stimulated mPTP opening. Treatment of SMNKO mice with the Cyclophilin D inhibitor, Cyclosporine A, increased membrane integrity, decreased the number of centralized nuclei, and increased survival. Conclusions Our study demonstrates that NAMPT is crucial for maintaining cellular Ca2+ homeostasis and skeletal muscle development, which is vital for juvenile survival. NAD+ salvage capacity is important for skeletal muscle development and survival. Skeletal muscle-specific Nampt knockout mice exhibit a dystrophy-like phenotype. Nampt deletion alters Ca2+ homeostasis and impairs mitochondrial function. Low NAD+ levels signals mitochondrial permeability transition pore opening. Cyclosporin A treatment improves sarcolemma integrity and increases survival rate.
Collapse
Affiliation(s)
- Astrid L Basse
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marianne Agerholm
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jean Farup
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark; Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Emilie Dalbram
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Joachim Nielsen
- Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
| | - Niels Ørtenblad
- Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
| | - Ali Altıntaş
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Amy M Ehrlich
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Krag
- Copenhagen Neuromuscular Center, Rigshospitalet, Copenhagen, Denmark
| | - Santina Bruzzone
- Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Morten Dall
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Roldan M de Guia
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jonas B Jensen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Andreas B Møller
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Anders Karlsen
- Institute of Sports Medicine, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Michael Kjær
- Institute of Sports Medicine, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Romain Barrès
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - John Vissing
- Copenhagen Neuromuscular Center, Rigshospitalet, Copenhagen, Denmark
| | - Steen Larsen
- Center for Healthy Aging, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Niels Jessen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark; Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
34
|
Germline Genetic Association between Stromal Interaction Molecule 1 (STIM1) and Clinical Outcomes in Breast Cancer Patients. J Pers Med 2020; 10:jpm10040287. [PMID: 33348924 PMCID: PMC7767290 DOI: 10.3390/jpm10040287] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/09/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022] Open
Abstract
Among all cancers in women, breast cancer has the highest incidence. The mortality of breast cancer is highly associated with metastasis. Migration and malignant transformation of cancer cells have been reported to be modulated by store-operated calcium (SOC) channels, which control calcium signaling and cell proliferation pathways. Stromal interaction molecule 1 (STIM1) is a calcium sensor in the endoplasmic reticulum, triggering the activation of store-operated calcium signaling. However, the clinical relevance of STIM1 in breast cancer is still unclear. Here, we recruited 348 breast cancer patients and conducted a genetic association study to address this question. Four tagging germline single nucleotide variants (SNVs) in STIM1 were selected and RNA sequencing data of 525 breast cancer samples from The Cancer Genome Atlas (TCGA) database were evaluated. The results show that rs2304891 and rs3750996 were correlated with clinical stage of breast cancer. Expression quantitative trait loci (eQTL) analysis indicated that risk G allele of STIM1 contributed to the higher expression of STIM1. In addition, we found an increased risk of rs2304891 G allele and rs3750996 A allele in estrogen receptor (ER) positive and progesterone receptor (PR) positive patients. In conclusion, our results suggest that germline SNV, rs2304891 and rs3750996 as well as STIM1 expression are important biomarkers for the prediction of clinical outcomes in breast cancer patients.
Collapse
|
35
|
Sun J, Chen F, Chen C, Zhang Z, Zhang Z, Tian W, Yu J, Wang K. Intestinal mRNA expression profile and bioinformatics analysis in a methamphetamine-induced mouse model of inflammatory bowel disease. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1669. [PMID: 33490181 PMCID: PMC7812166 DOI: 10.21037/atm-20-7741] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Background Methamphetamine use has become a serious global public health problem and puts increasing burdens on healthcare services. Abdominal complications caused by methamphetamine use are uncommon and often go ignored by clinicians. The exact intestinal pathological alterations and transcriptomic responses associated with methamphetamine use are not well understood. This study sought to investigate the transcriptome in a methamphetamine-induced mouse model of inflammatory bowel disease (IBD) using next-generation RNA sequencing. Methods Tissues from the ileum of methamphetamine-treated mice (n=5) and control mice (n=5) were dissected, processed and applied to RNA-sequencing. Bioinformatics and histopathological analysis were then performed. The expression profiles of intestinal tissue samples were analyzed and their expression profiles were integrated to obtain the differentially expressed genes and analyzed using bioinformatics. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses of the differentially expressed genes were performed using Metascape. Results A total of 326 differentially expressed genes were identified; of these genes, 120 were upregulated and 206 were downregulated. The Gene Ontology analysis showed that the biological processes of the differentially expressed genes were focused primarily on the regulation of cellular catabolic processes, endocytosis, and autophagy. The main cellular components included the endoplasmic and endocytic vesicles, cytoskeleton, adherens junctions, focal adhesions, cell body, and lysosomes. Molecular functions included protein transferase, GTPase and proteinase activities, actin-binding, and protein-lipid complex binding. The Kyoto Encyclopedia of Genes and Genomes pathway analysis showed that the differentially expressed genes were mainly involved in bacterial invasion of epithelial cells, protein processing in the endoplasmic reticulum, regulation of the actin cytoskeleton, and T-cell receptor signaling pathways. A set of overlapping genes between IBD and methamphetamine-treated intestinal tissues was discovered. Conclusions The present study is the first to analyze intestinal samples from methamphetamine-treated mice using high-throughput RNA sequencing. This study revealed key molecules that might be involved in the pathogenesis of a special type of methamphetamine-induced IBD. These results offer new insights into the relationship between methamphetamine abuse and IBD.
Collapse
Affiliation(s)
- Jiaxue Sun
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China.,Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, China.,The Scientific Research Laboratory Center, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Fengrong Chen
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China.,The Scientific Research Laboratory Center, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Cheng Chen
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China.,Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, China.,The Scientific Research Laboratory Center, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zherui Zhang
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China.,Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, China.,The Scientific Research Laboratory Center, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zunyue Zhang
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China.,The Scientific Research Laboratory Center, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Weiwei Tian
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China.,The Scientific Research Laboratory Center, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Juehua Yu
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China.,The Scientific Research Laboratory Center, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Kunhua Wang
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
36
|
Du M, Wang X, Yuan L, Liu B, Mao X, Huang D, Yang L, Huang K, Zhang F, Wang Y, Luo X, Wang C, Peng J, Liang M, Huang D, Huang K. Targeting NFATc4 attenuates non-alcoholic steatohepatitis in mice. J Hepatol 2020; 73:1333-1346. [PMID: 32717288 DOI: 10.1016/j.jhep.2020.07.030] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 06/22/2020] [Accepted: 07/16/2020] [Indexed: 02/09/2023]
Abstract
BACKGROUND & AIMS The nuclear factor of activated T-cells (NFAT) family was first recognised to play an important role in the differentiation of T cells, but has since been shown to regulate multiple pathophysiological processes. However, whether it is involved in the pathogenesis of non-alcoholic steatohepatitis (NASH) remains unknown. METHODS Hepatic NFATc expression and localisation were analysed in C57BL/6 mice on a methionine-choline-deficient diet, as well as in samples from non-alcoholic fatty liver disease patients. Gain- or loss-of-function approaches were used to investigate the role of NFATc4 in NASH. RESULTS NFATc4 translocates from the cytoplasm to the nucleus in hepatocytes of both humans and rodents with NASH. NFATc4 knockdown resulted in decreased hepatic steatosis, inflammation, and fibrosis during NASH progression. Mechanistically, we found that activated NFATc4 directly bound to peroxisome proliferator-activated receptor α (PPARα) in the nucleus and negatively regulated its transcriptional activity, thereby impairing the hepatic fatty acid oxidation pathway and increasing lipid deposition in the liver. Moreover, NFATc4 activation increased the production and secretion of osteopontin (OPN) from hepatocytes, which subsequently enhanced the macrophage-mediated inflammatory response and hepatic stellate cell-mediated fibrosis progression via paracrine signalling. CONCLUSIONS Hepatic NFATc4 activation accelerates the progression of NASH by suppressing PPARα signalling and increasing OPN expression. Genetic or pharmacological inhibition of NFATc4 may have potential for future therapy of NASH. LAY SUMMARY NFATc4 is activated in the non-alcoholic steatohepatitis of mice and patients. Inhibition of NFATc4 activation alleviates lipid deposition, inflammatory response, and fibrosis progression in the liver.
Collapse
Affiliation(s)
- Meng Du
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xiaojing Wang
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Yuan
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bing Liu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxiang Mao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dandan Huang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liu Yang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kun Huang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fengxiao Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Wang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xi Luo
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cheng Wang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiangtong Peng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Minglu Liang
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dan Huang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Huang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
37
|
Jinqiang Zhuang MD, Ruijun Yuan MD, Yizeng MD, Congliang Miao MD, Dandan Zhou MD, Anli Na MD, Xinying Yang MD, Hui Xu MD, Hong J. The CnB1 p.D102A variant is linked to dilated cardiomyopathy via impaired Calcineurin activity. J Mol Cell Cardiol 2020; 148:15-24. [PMID: 32882262 DOI: 10.1016/j.yjmcc.2020.08.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/30/2020] [Accepted: 08/18/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND The role of calcineurin (protein phosphatase 2B (PP2B)) in the pathogenesis of human dilated cardiomyopathy (DCM) has not been fully elucidated. We determined the potential involvement of calcineurin in the pathogenesis of DCM caused by mutations in CnB1, a subunit of calcineurin. METHODS By whole-exome sequencing, we identified a new CnB1 variant in a Han Chinese proband with cardiomyopathy from a 3-generation family with 2 normal individuals and 3 individuals with familial dilated cardiomyopathy. The potential pathogenic variant was validated by Sanger sequencing. We performed functional and mechanistic experiments in a CnB1-knockin (KI) mouse model and at the cellular level. RESULTS We detected a rare heterozygous CnB1 variant (p.D102A) in a proband with dilated cardiomyopathy. This variant was localized to the EF hand 3 region of CnB1, where no variants have been previously reported. KI mice harboring the p.D102A variant exhibited decreased cardiac function and cardiac dilatation. Immunoblotting, RT-PCR and immunofluorescence results showed decreased cardiomyocyte size and heart failure-related protein expression. A calcineurin activity assay demonstrated decreased calcineurin activity in the KI mice, accompanied by the decreased ability of CnB1 to bind CnA. CONCLUSIONS CnB1 p.D102A is a disease-associated variant that confers susceptibility to cardiac dilatation. This variant is associated with impaired calcineurin activity and a subsequent decrease in the ability of CnB1 to bind CnA.
Collapse
Affiliation(s)
- M D Jinqiang Zhuang
- Department of Internal and Emergency Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China
| | - M D Ruijun Yuan
- Department of Cardiovascular Surgery, Changhai Hospital, Second Military Medical University. Shanghai, China
| | - M D Yizeng
- Department of Critical Care Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Shangcheng District, Hangzhou, 310003, China
| | - M D Congliang Miao
- Department of Internal and Emergency Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China
| | - M D Dandan Zhou
- Department of Internal and Emergency Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China
| | - M D Anli Na
- Department of Internal and Emergency Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China
| | - M D Xinying Yang
- Department of Internal and Emergency Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China
| | - M D Hui Xu
- Department of Internal and Emergency Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China
| | - Jiang Hong
- Department of Internal and Emergency Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China.
| |
Collapse
|
38
|
Vesentini G, Barbosa AMP, Floriano JF, Felisbino SL, Costa SMB, Piculo F, Marini G, Nunes SK, Reyes DRA, Marcondes JPC, Hallur RLS, Rozza AL, Magalhães CG, Costa R, Abbade JF, Corrente JE, Calderon IMP, Matheus SMM, Rudge MVC. Deleterious effects of gestational diabetes mellitus on the characteristics of the rectus abdominis muscle associated with pregnancy-specific urinary incontinence. Diabetes Res Clin Pract 2020; 166:108315. [PMID: 32679058 DOI: 10.1016/j.diabres.2020.108315] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 06/05/2020] [Accepted: 07/07/2020] [Indexed: 12/15/2022]
Abstract
AIMS To evaluate the effects of gestational diabetes mellitus (GDM) on the structural characteristics of the rectus abdominis muscle (RAM) and its indirect effects on pregnancy-specific urinary incontinence (PSUI). METHODS A total of 92 pregnant women were divided into four groups, according to their clinical conditions: non-GDM continent, non-GDM associated PSUI, GDM continent and GDM associated PSUI. The muscle morphometry (histochemistry and immunohistochemistry) for the fiber types and collagen fiber distribution, the ultrastructural analysis (transmission electron microscopy), the protein expression of fiber types and calcium signaling (Western blotting), and the content of types I and III collagen fiber (ELISA) in RAM collected at delivery were assessed. RESULTS The GDM groups presented a significantly increased number of slow fibers and slow-twitch oxidative fiber expression; decreased fiber area, number of fast fibers, and area of collagen; an increase in central nuclei; ultrastructural alterations with focal lesion areas such as myeloid structures, sarcomere disorganization, and mitochondrial alteration. The PSUI groups presented a considerable decrease in types I and III collagen contents and the localization of collagen fiber. CONCLUSIONS Our data reveal that GDM causes morphological, biochemical and physiological changes in the RAM, and this might predispose women to PSUI.
Collapse
Affiliation(s)
- Giovana Vesentini
- Perinatal Diabetes Research Center, University Hospital, Botucatu Medical School, Univ Estadual Paulista_UNESP, São Paulo State, Brazil; São Paulo State University (UNESP), Botucatu Medical School, Department of Gynecology and Obstetrics, Botucatu, Sao Paulo, Brazil
| | - Angélica M P Barbosa
- São Paulo State University (UNESP), School of Philosophy and Sciences, Department of Physical Therapy and Occupational Therapy, Marilia, São Paulo State, Brazil
| | - Juliana F Floriano
- Perinatal Diabetes Research Center, University Hospital, Botucatu Medical School, Univ Estadual Paulista_UNESP, São Paulo State, Brazil; São Paulo State University (UNESP), Botucatu Medical School, Department of Gynecology and Obstetrics, Botucatu, Sao Paulo, Brazil
| | - Sérgio L Felisbino
- São Paulo State University (UNESP), Institute of Biosciences, Department of Morphology, Botucatu, São Paulo State, Brazil
| | - Sarah M B Costa
- Perinatal Diabetes Research Center, University Hospital, Botucatu Medical School, Univ Estadual Paulista_UNESP, São Paulo State, Brazil; São Paulo State University (UNESP), Botucatu Medical School, Department of Gynecology and Obstetrics, Botucatu, Sao Paulo, Brazil
| | - Fernanda Piculo
- Perinatal Diabetes Research Center, University Hospital, Botucatu Medical School, Univ Estadual Paulista_UNESP, São Paulo State, Brazil; São Paulo State University (UNESP), Botucatu Medical School, Department of Gynecology and Obstetrics, Botucatu, Sao Paulo, Brazil
| | - Gabriela Marini
- Perinatal Diabetes Research Center, University Hospital, Botucatu Medical School, Univ Estadual Paulista_UNESP, São Paulo State, Brazil; São Paulo State University (UNESP), Botucatu Medical School, Department of Gynecology and Obstetrics, Botucatu, Sao Paulo, Brazil; Universidade Sagrado Coração, Department of Health Sciences, Bauru, São Paulo, Brazil
| | - Sthefanie K Nunes
- Perinatal Diabetes Research Center, University Hospital, Botucatu Medical School, Univ Estadual Paulista_UNESP, São Paulo State, Brazil; São Paulo State University (UNESP), Botucatu Medical School, Department of Gynecology and Obstetrics, Botucatu, Sao Paulo, Brazil
| | - David R A Reyes
- Perinatal Diabetes Research Center, University Hospital, Botucatu Medical School, Univ Estadual Paulista_UNESP, São Paulo State, Brazil; São Paulo State University (UNESP), Botucatu Medical School, Department of Gynecology and Obstetrics, Botucatu, Sao Paulo, Brazil
| | - João P C Marcondes
- Perinatal Diabetes Research Center, University Hospital, Botucatu Medical School, Univ Estadual Paulista_UNESP, São Paulo State, Brazil; São Paulo State University (UNESP), Botucatu Medical School, Department of Gynecology and Obstetrics, Botucatu, Sao Paulo, Brazil
| | - Raghavendra L S Hallur
- Perinatal Diabetes Research Center, University Hospital, Botucatu Medical School, Univ Estadual Paulista_UNESP, São Paulo State, Brazil; São Paulo State University (UNESP), Botucatu Medical School, Department of Gynecology and Obstetrics, Botucatu, Sao Paulo, Brazil
| | - Ariane L Rozza
- São Paulo State University (UNESP), Institute of Biosciences, Department of Morphology, Botucatu, São Paulo State, Brazil
| | - Cláudia G Magalhães
- Perinatal Diabetes Research Center, University Hospital, Botucatu Medical School, Univ Estadual Paulista_UNESP, São Paulo State, Brazil; São Paulo State University (UNESP), Botucatu Medical School, Department of Gynecology and Obstetrics, Botucatu, Sao Paulo, Brazil
| | - Roberto Costa
- Perinatal Diabetes Research Center, University Hospital, Botucatu Medical School, Univ Estadual Paulista_UNESP, São Paulo State, Brazil; São Paulo State University (UNESP), Botucatu Medical School, Department of Gynecology and Obstetrics, Botucatu, Sao Paulo, Brazil
| | - Joelcio F Abbade
- Perinatal Diabetes Research Center, University Hospital, Botucatu Medical School, Univ Estadual Paulista_UNESP, São Paulo State, Brazil; São Paulo State University (UNESP), Botucatu Medical School, Department of Gynecology and Obstetrics, Botucatu, Sao Paulo, Brazil
| | - José E Corrente
- São Paulo State University (UNESP), Institute of Biosciences, Biostatistics Department, Botucatu, São Paulo, Brazil
| | - Iracema M P Calderon
- Perinatal Diabetes Research Center, University Hospital, Botucatu Medical School, Univ Estadual Paulista_UNESP, São Paulo State, Brazil; São Paulo State University (UNESP), Botucatu Medical School, Department of Gynecology and Obstetrics, Botucatu, Sao Paulo, Brazil
| | - Selma M M Matheus
- São Paulo State University (UNESP), Institute of Biosciences, Department of Anatomy, Botucatu, São Paulo State, Brazil
| | - Marilza V C Rudge
- Perinatal Diabetes Research Center, University Hospital, Botucatu Medical School, Univ Estadual Paulista_UNESP, São Paulo State, Brazil; São Paulo State University (UNESP), Botucatu Medical School, Department of Gynecology and Obstetrics, Botucatu, Sao Paulo, Brazil.
| |
Collapse
|
39
|
Elbaz M, Ruiz A, Nicolay S, Tupini C, Bachmann C, Eckhardt J, Benucci S, Pelczar P, Treves S, Zorzato F. Bi-allelic expression of the RyR1 p.A4329D mutation decreases muscle strength in slow-twitch muscles in mice. J Biol Chem 2020; 295:10331-10339. [PMID: 32499372 DOI: 10.1074/jbc.ra120.013846] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 05/29/2020] [Indexed: 12/25/2022] Open
Abstract
Mutations in the ryanodine receptor 1 (RYR1) gene are associated with several human congenital myopathies, including the dominantly inherited central core disease and exercise-induced rhabdomyolysis, and the more severe recessive phenotypes, including multiminicore disease, centronuclear myopathy, and congenital fiber type disproportion. Within the latter group, those carrying a hypomorphic mutation in one allele and a missense mutation in the other are the most severely affected. Because of nonsense-mediated decay, most hypomorphic alleles are not expressed, resulting in homozygous expression of the missense mutation allele. This should result in 50% reduced expression of the ryanodine receptor in skeletal muscle, but its observed content is even lower. To study in more detail the biochemistry and pathophysiology of recessive RYR1 myopathies, here we investigated a mouse model we recently generated by analyzing the effect of bi-allelic versus mono-allelic expression of the RyR1 p.A4329D mutation. Our results revealed that the expression of two alleles carrying the same mutation or of one allele with the mutation in combination with a hypomorphic allele does not result in functionally equal outcomes and impacts skeletal muscles differently. In particular, the bi-allelic RyR1 p.A4329D mutation caused a milder phenotype than its mono-allelic expression, leading to changes in the biochemical properties and physiological function only of slow-twitch muscles and largely sparing fast-twitch muscles. In summary, bi-allelic expression of the RyR1 p.A4329D mutation phenotypically differs from mono-allelic expression of this mutation in a compound heterozygous carrier.
Collapse
Affiliation(s)
- Moran Elbaz
- Department of Biomedicine, Basel University Hospital, Basel, Switzerland
| | - Alexis Ruiz
- Department of Biomedicine, Basel University Hospital, Basel, Switzerland
| | - Sven Nicolay
- Department of Biomedicine, Basel University Hospital, Basel, Switzerland
| | - Chiara Tupini
- Department of Life Science and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Christoph Bachmann
- Department of Biomedicine, Basel University Hospital, Basel, Switzerland
| | - Jan Eckhardt
- Department of Biomedicine, Basel University Hospital, Basel, Switzerland
| | - Sofia Benucci
- Department of Biomedicine, Basel University Hospital, Basel, Switzerland
| | - Pawel Pelczar
- Center for Transgenic Models, University of Basel, Basel, Switzerland
| | - Susan Treves
- Department of Biomedicine, Basel University Hospital, Basel, Switzerland.,Department of Life Science and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Francesco Zorzato
- Department of Biomedicine, Basel University Hospital, Basel, Switzerland .,Department of Life Science and Biotechnology, University of Ferrara, Ferrara, Italy
| |
Collapse
|
40
|
Sharma B, Dabur R. Role of Pro-inflammatory Cytokines in Regulation of Skeletal Muscle Metabolism: A Systematic Review. Curr Med Chem 2020; 27:2161-2188. [DOI: 10.2174/0929867326666181129095309] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 11/15/2018] [Accepted: 11/19/2018] [Indexed: 12/18/2022]
Abstract
Background:
Metabolic pathways perturbations lead to skeletal muscular atrophy in the
cachexia and sarcopenia due to increased catabolism. Pro-inflammatory cytokines induce the catabolic
pathways that impair the muscle integrity and function. Hence, this review primarily concentrates
on the effects of pro-inflammatory cytokines in regulation of skeletal muscle metabolism.
Objective:
This review will discuss the role of pro-inflammatory cytokines in skeletal muscles during
muscle wasting conditions. Moreover, the coordination among the pro-inflammatory cytokines
and their regulated molecular signaling pathways which increase the protein degradation will be
discussed.
Results:
During normal conditions, pro-inflammatory cytokines are required to balance anabolism
and catabolism and to maintain normal myogenesis process. However, during muscle wasting their
enhanced expression leads to marked destructive metabolism in the skeletal muscles. Proinflammatory
cytokines primarily exert their effects by increasing the expression of calpains and E3
ligases as well as of Nf-κB, required for protein breakdown and local inflammation. Proinflammatory
cytokines also locally suppress the IGF-1and insulin functions, hence increase the
FoxO activation and decrease the Akt function, the central point of carbohydrates lipid and protein
metabolism.
Conclusion:
Current advancements have revealed that the muscle mass loss during skeletal muscular
atrophy is multifactorial. Despite great efforts, not even a single FDA approved drug is available
in the market. It indicates the well-organized coordination among the pro-inflammatory cytokines
that need to be further understood and explored.
Collapse
Affiliation(s)
- Bhawana Sharma
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, Haryana-124001, India
| | - Rajesh Dabur
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, Haryana-124001, India
| |
Collapse
|
41
|
The role of Ca 2+/NFAT in Dysfunction and Inflammation of Human Coronary Endothelial Cells induced by Sera from patients with Kawasaki disease. Sci Rep 2020; 10:4706. [PMID: 32170198 PMCID: PMC7069934 DOI: 10.1038/s41598-020-61667-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 02/26/2020] [Indexed: 12/14/2022] Open
Abstract
Ca2+/nuclear factor of activated T-cells (Ca2+/NFAT) signaling pathway may play a crucial role in the pathogenesis of Kawasaki disease (KD). We investigated the poorly understood Ca2+/NFAT regulation of coronary artery endothelial cells and consequent dysfunction in KD pathogenesis. Human coronary artery endothelial cells (HCAECs) stimulated with sera from patients with KD, compared with sera from healthy children, exhibited significant increases in proliferation and angiogenesis, higher levels of NFATc1 and NFATc3 and some inflammatory molecules, and increased nuclear translocation of NFATc1 and NFATc3. HCAECs stimulated with sera from patients with KD treated with cyclosporine A (CsA) showed decreased proliferation, angiogenesis, NFATc1 and inflammatory molecules levels as compared with results for untreated HCAECs. In conclusion, our data reveal that KD sera activate the Ca2+/NFAT in HCAECs, leading to dysfunction and inflammation of endothelial cells. CsA has cytoprotective effects by ameliorating endothelial cell homeostasis via Ca2+/NFAT.
Collapse
|
42
|
Petersen CE, Wolf MJ, Smyth JT. Suppression of store-operated calcium entry causes dilated cardiomyopathy of the Drosophila heart. Biol Open 2020; 9:bio049999. [PMID: 32086252 PMCID: PMC7075072 DOI: 10.1242/bio.049999] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 02/07/2020] [Indexed: 11/20/2022] Open
Abstract
Store-operated Ca2+ entry (SOCE) is an essential Ca2+ signaling mechanism present in most animal cells. SOCE refers to Ca2+ influx that is activated by depletion of sarco/endoplasmic reticulum (S/ER) Ca2+ stores. The main components of SOCE are STIM and Orai. STIM proteins function as S/ER Ca2+ sensors, and upon S/ER Ca2+ depletion STIM rearranges to S/ER-plasma membrane junctions and activates Orai Ca2+ influx channels. Studies have implicated SOCE in cardiac hypertrophy pathogenesis, but SOCE's role in normal heart physiology remains poorly understood. We therefore analyzed heart-specific SOCE function in Drosophila, a powerful animal model of cardiac physiology. We show that heart-specific suppression of Stim and Orai in larvae and adults resulted in reduced contractility consistent with dilated cardiomyopathy. Myofibers were also highly disorganized in Stim and Orai RNAi hearts, reflecting possible decompensation or upregulated stress signaling. Furthermore, we show that reduced heart function due to SOCE suppression adversely affected animal viability, as heart specific Stim and Orai RNAi animals exhibited significant delays in post-embryonic development and adults died earlier than controls. Collectively, our results demonstrate that SOCE is essential for physiological heart function, and establish Drosophila as an important model for understanding the role of SOCE in cardiac pathophysiology.
Collapse
Affiliation(s)
- Courtney E Petersen
- Graduate Program in Molecular and Cellular Biology, Uniformed Services University of the Health Sciences, F. Edward Hébert School of Medicine, Bethesda, MD 20814, USA
| | - Matthew J Wolf
- Division of Cardiovascular Medicine, Department of Medicine, The University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Jeremy T Smyth
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, F. Edward Hébert School of Medicine, Bethesda, MD 20814, USA
| |
Collapse
|
43
|
Park HS, Lee SC, Cardenas ME, Heitman J. Calcium-Calmodulin-Calcineurin Signaling: A Globally Conserved Virulence Cascade in Eukaryotic Microbial Pathogens. Cell Host Microbe 2020; 26:453-462. [PMID: 31600499 DOI: 10.1016/j.chom.2019.08.004] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 06/29/2019] [Accepted: 08/02/2019] [Indexed: 12/19/2022]
Abstract
Calcium is an abundant intracellular ion, and calcium homeostasis plays crucial roles in several cellular processes. The calcineurin signaling cascade is one of the major pathways governed by intracellular calcium. Calcineurin, a conserved protein from yeast to humans, is a calcium-calmodulin-dependent serine-threonine-specific phosphatase that orchestrates cellular stress responses. In eukaryotic microbial pathogens, calcineurin controls essential virulence pathways, such as the ability to grow at host temperature, morphogenesis to enable invasive hyphal growth, drug tolerance and resistance, cell wall integrity, and sexual development. Therefore, the calcineurin cascade is an attractive target in drug development against eukaryotic pathogens. In the present review, we summarize and discuss the current knowledge on the roles of calcineurin in eukaryotic microbial pathogens, focusing on fungi and parasitic protists.
Collapse
Affiliation(s)
- Hee-Soo Park
- School of Food Science and Biotechnology, Institute of Agricultural Science and Technology, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Soo Chan Lee
- South Texas Center for Emerging Infectious Diseases (STCEID), Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Maria E Cardenas
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Joseph Heitman
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
44
|
Kim HM, He L, Lee S, Park C, Kim DH, Han HJ, Han J, Hwang J, Cha-Molstad H, Lee KH, Ko SK, Jang JH, Ryoo IJ, Blenis J, Lee HG, Ahn JS, Kwon YT, Soung NK, Kim BY. Inhibition of osteoclasts differentiation by CDC2-induced NFATc1 phosphorylation. Bone 2020; 131:115153. [PMID: 31730830 DOI: 10.1016/j.bone.2019.115153] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/18/2019] [Accepted: 11/11/2019] [Indexed: 10/25/2022]
Abstract
Bone homeostasis is regulated by a balance of bone formation and bone resorption; dysregulation of bone homeostasis may cause bone-related diseases (e.g., osteoporosis, osteopetrosis, bone fracture). Members of the nuclear factor of activated T cells (NFAT) family of transcription factors play crucial roles in the regulation of immune system, inflammatory responses, cardiac formation, skeletal muscle development, and bone homeostasis. Of these, NFATc1 is a key transcription factor mediating osteoclast differentiation, which is regulated by phosphorylation by distinct NFAT kinases including casein kinase 1 (CK1), glycogen synthase kinase 3 (GSK3), and dual-specificity tyrosine-phosphorylation-regulated kinases (DYRKs). In this study, we report that cell division control protein 2 homolog (cdc2) is a novel NFAT protein kinase that inhibits NFATc1 activation by direct phosphorylation of the NFATc1 S263 residue. Cdc2 inhibitors such as Roscovitine and BMI-1026 induce reduction of phosphorylation of NFATc1, and this process leads to the inhibition of NFATc1 translocation from the nucleus to the cytoplasm, consequently increasing the nuclear pool of NFATc1. Additionally, the inhibition of cdc2-mediated NFATc1 phosphorylation causes an elevation of osteoclast differentiation or TRAP-positive staining in zebrafish scales. Our results suggest that cdc2 is a novel NFAT protein kinase that negatively regulates osteoclast differentiation.
Collapse
Affiliation(s)
- Hye-Min Kim
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea
| | - Long He
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea; Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA
| | - Sangku Lee
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea
| | - Chanmi Park
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea
| | - Dong Hyun Kim
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea; Department of Biomolecular Science, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Ho-Jin Han
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea; Department of Biomolecular Science, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Junyeol Han
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea; Department of Biomolecular Science, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Joonsung Hwang
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea
| | - Hyunjoo Cha-Molstad
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea
| | - Kyung Ho Lee
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea
| | - Sung-Kyun Ko
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea
| | - Jae-Hyuk Jang
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea
| | - In-Ja Ryoo
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea
| | - John Blenis
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA
| | - Hee Gu Lee
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Jong Seog Ahn
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea; Department of Biomolecular Science, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Yong Tae Kwon
- Protein Metabolism Medical Research Center, Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea.
| | - Nak-Kyun Soung
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea; Department of Biomolecular Science, University of Science and Technology, Daejeon 34113, Republic of Korea.
| | - Bo Yeon Kim
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea; Department of Biomolecular Science, University of Science and Technology, Daejeon 34113, Republic of Korea.
| |
Collapse
|
45
|
Innes AQ, Thomson G, Cotter M, King JA, Vollaard NBJ, Kelly BM. Evaluating differences in the clinical impact of a free online weight loss programme, a resource-intensive commercial weight loss programme and an active control condition: a parallel randomised controlled trial. BMC Public Health 2019; 19:1732. [PMID: 31870345 PMCID: PMC6929498 DOI: 10.1186/s12889-019-8061-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 12/11/2019] [Indexed: 12/22/2022] Open
Abstract
Background Finding effective intervention strategies to combat rising obesity levels could significantly reduce the burden that obesity and associated non-communicable diseases places on both individuals and the National Health Service. Methods In this parallel randomised-controlled trial, 76 participants who are overweight or obese (50 female) were given free access to a fitness centre for the duration of the 12-week intervention and randomised to one of three interventions. The commercial intervention, the Healthy Weight Programme, (HWP, n = 25, 10/15 men/women) consisted of twelve 1-h nutrition coaching sessions with a nutritionist delivered as a mixture of group and 1 to 1 sessions. In addition, twice-weekly exercise sessions (24 in total) were delivered by personal trainers for 12 weeks. The NHS intervention (n = 25, 8/17 men/women) consisted of following an entirely self-managed 12-week online NHS resource. The GYM intervention (n = 26, 8/18 men/women) received no guidance or formal intervention. All participants were provided with a gym induction for safety and both the NHS and GYM participants were familiarised with ACSM physical activity guidelines by way of a hand-out. Results The overall follow-up rate was 83%. Body mass was significantly reduced at post-intervention in all groups (HWP: N = 18, − 5.17 ± 4.22 kg, NHS: N = 21–4.19 ± 5.49 kg; GYM: N = 24–1.17 ± 3.00 kg; p < 0.001) with greater reductions observed in HWP and NHS groups compared to GYM (p < 0.05). Out with body mass and BMI, there were no additional statistically significant time x intervention interaction effects. Conclusions This is the first study to evaluate the efficacy of both a free online NHS self-help weight-loss tool and a commercial weight loss programme that provides face-to-face nutritional support and supervised exercise. The findings suggest that both interventions are superior to an active control condition with regard to eliciting short-term weight-loss. Trial registration ISRCTN Registry - ISRCTN31489026. Prospectively registered: 27/07/16.
Collapse
Affiliation(s)
- Aidan Q Innes
- Nuffield Health Research Group, Nuffield Health, Ashley Avenue, Epsom, Surrey, KT18 5AL, UK.,Faculty of Health, Psychology and Social Care, Manchester Metropolitan University, Manchester, M15 6GX, UK
| | - Greig Thomson
- Nuffield Health Research Group, Nuffield Health, Ashley Avenue, Epsom, Surrey, KT18 5AL, UK
| | - Mary Cotter
- Nuffield Health Research Group, Nuffield Health, Ashley Avenue, Epsom, Surrey, KT18 5AL, UK
| | - James A King
- National Centre for Sport and Exercise Medicine, Loughborough University, Loughborough, LE11 3TU, UK.,National Institute for Health Research Leicester Biomedical Research Centre, Leicester, LE3 8QD, UK
| | - Niels B J Vollaard
- Faculty of Health Sciences and Sport, University of Stirling, Stirling, FK9 4LA, UK
| | - Benjamin M Kelly
- Nuffield Health Research Group, Nuffield Health, Ashley Avenue, Epsom, Surrey, KT18 5AL, UK. .,Faculty of Health, Psychology and Social Care, Manchester Metropolitan University, Manchester, M15 6GX, UK.
| |
Collapse
|
46
|
Pickering C, Suraci B, Semenova EA, Boulygina EA, Kostryukova ES, Kulemin NA, Borisov OV, Khabibova SA, Larin AK, Pavlenko AV, Lyubaeva EV, Popov DV, Lysenko EA, Vepkhvadze TF, Lednev EM, Leońska-Duniec A, Pająk B, Chycki J, Moska W, Lulińska-Kuklik E, Dornowski M, Maszczyk A, Bradley B, Kana-Ah A, Cięszczyk P, Generozov EV, Ahmetov II. A Genome-Wide Association Study of Sprint Performance in Elite Youth Football Players. J Strength Cond Res 2019; 33:2344-2351. [PMID: 31343553 DOI: 10.1519/jsc.0000000000003259] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Pickering, C, Suraci, B, Semenova, EA, Boulygina, EA, Kostryukova, ES, Kulemin, NA, Borisov, OV, Khabibova, SA, Larin, AK, Pavlenko, AV, Lyubaeva, EV, Popov, DV, Lysenko, EA, Vepkhvadze, TF, Lednev, EM, Leońska-Duniec, A, Pająk, B, Chycki, J, Moska, W, Lulińska-Kuklik, E, Dornowski, M, Maszczyk, A, Bradley, B, Kana-ah, A, Cięszczyk, P, Generozov, EV, and Ahmetov, II. A genome-wide association study of sprint performance in elite youth football players. J Strength Cond Res 33(9): 2344-2351, 2019-Sprint speed is an important component of football performance, with teams often placing a high value on sprint and acceleration ability. The aim of this study was to undertake the first genome-wide association study to identify genetic variants associated with sprint test performance in elite youth football players and to further validate the obtained results in additional studies. Using micro-array data (600 K-1.14 M single nucleotide polymorphisms [SNPs]) of 1,206 subjects, we identified 12 SNPs with suggestive significance after passing replication criteria. The polymorphism rs55743914 located in the PTPRK gene was found as the most significant for 5-m sprint test (p = 7.7 × 10). Seven of the discovered SNPs were also associated with sprint test performance in a cohort of 126 Polish women, and 4 were associated with power athlete status in a cohort of 399 elite Russian athletes. Six SNPs were associated with muscle fiber type in a cohort of 96 Russian subjects. We also examined genotype distributions and possible associations for 16 SNPs previously linked with sprint performance. Four SNPs (AGT rs699, HSD17B14 rs7247312, IGF2 rs680, and IL6 rs1800795) were associated with sprint test performance in this cohort. In addition, the G alleles of 2 SNPs in ADRB2 (rs1042713 & rs1042714) were significantly over-represented in these players compared with British and European controls. These results suggest that there is a genetic influence on sprint test performance in footballers, and identifies some of the genetic variants that help explain this influence.
Collapse
Affiliation(s)
- Craig Pickering
- School of Sport and Wellbeing, Institute of Coaching and Performance, University of Central Lancashire, Preston, United Kingdom.,Prenetics DNAFit Research Center, London, United Kingdom
| | - Bruce Suraci
- Academy Coaching Department, AFC Bournemouth, Bournemouth, United Kingdom
| | - Ekaterina A Semenova
- Department of Molecular Biology and Genetics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia.,Department of Biochemistry, Kazan Federal University, Kazan, Russia
| | | | - Elena S Kostryukova
- Department of Molecular Biology and Genetics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Nickolay A Kulemin
- Department of Molecular Biology and Genetics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Oleg V Borisov
- Department of Molecular Biology and Genetics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia.,Moscow Institute of Physics and Technology (State University), Moscow, Russia
| | - Sofya A Khabibova
- Department of Molecular Biology and Genetics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Andrey K Larin
- Department of Molecular Biology and Genetics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Alexander V Pavlenko
- Department of Molecular Biology and Genetics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Ekaterina V Lyubaeva
- Laboratory of Exercise Physiology, Institute of Biomedical Problems of the Russian Academy of Sciences, Moscow, Russia
| | - Daniil V Popov
- Laboratory of Exercise Physiology, Institute of Biomedical Problems of the Russian Academy of Sciences, Moscow, Russia
| | - Evgeny A Lysenko
- Laboratory of Exercise Physiology, Institute of Biomedical Problems of the Russian Academy of Sciences, Moscow, Russia
| | - Tatiana F Vepkhvadze
- Laboratory of Exercise Physiology, Institute of Biomedical Problems of the Russian Academy of Sciences, Moscow, Russia
| | - Egor M Lednev
- Laboratory of Exercise Physiology, Institute of Biomedical Problems of the Russian Academy of Sciences, Moscow, Russia
| | - Agata Leońska-Duniec
- Faculty of Physical Education, Gdansk University of Physical Education and Sport, Gdansk, Poland
| | - Beata Pająk
- Independent Laboratory of Genetics and Molecular Biology, Kaczkowski Military Institute of Hygiene Epidemiology, Poland
| | - Jakub Chycki
- Department of Sports Training, Academy of Physical Education, Katowice, Poland
| | - Waldemar Moska
- Faculty of Tourism and Recreation, Gdansk University of Physical Education and Sport, Gdansk, Poland
| | - Ewelina Lulińska-Kuklik
- Faculty of Tourism and Recreation, Gdansk University of Physical Education and Sport, Gdansk, Poland
| | - Marcin Dornowski
- Faculty of Physical Education, Gdansk University of Physical Education and Sport, Gdansk, Poland
| | - Adam Maszczyk
- Department of Theory and Practice of Sport, Academy of Physical Education in Katowice Poland
| | - Ben Bradley
- Academy Sports Science Department, AFC Bournemouth, Bournemouth, United Kingdom
| | - Adam Kana-Ah
- Academy Sports Science Department, AFC Bournemouth, Bournemouth, United Kingdom
| | - Pawel Cięszczyk
- Department of Theory and Practice of Sport, Academy of Physical Education in Katowice Poland
| | - Edward V Generozov
- Department of Molecular Biology and Genetics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Ildus I Ahmetov
- Department of Molecular Biology and Genetics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia.,Laboratory of Molecular Genetics, Kazan State Medical University, Kazan, Russia.,Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| |
Collapse
|
47
|
Hu J, Shi D, Ding M, Huang T, Gu R, Xiao J, Xian CJ, Dong J, Wang L, Liao H. Calmodulin-dependent signalling pathways are activated and mediate the acute inflammatory response of injured skeletal muscle. J Physiol 2019; 597:5161-5177. [PMID: 31506936 DOI: 10.1113/jp278478] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Accepted: 09/09/2019] [Indexed: 12/17/2022] Open
Abstract
KEY POINTS There is a close relationship between skeletal muscle physiology and Ca2+ /calmodulin (CaM) signalling. Despite the effects of Ca2+ /CaM signalling on immune and inflammatory responses having been extensively explored, few studies have investigated the role of CaM pathway activation on the post-injury muscle inflammatory response. In this study, we investigated the role of CaM-dependent signalling in muscle inflammation in cardiotoxin induced myoinjuries in mice. The Ca2+ /calmodulin-dependent protein kinase II (CaMII), Ca2+ /calmodulin-dependent protein kinase IV (CaMKIV), and nuclear factor of activated T cells (NFAT) pathways are likely to be simultaneously activated in muscle cells and in infiltrating lymphocytes and to regulate the immune behaviours of myofibres in an inflammatory environment, and these pathways ultimately affect the outcome of muscle inflammation. ABSTRACT Calcium/calmodulin (Ca2+ /CaM) signalling is essential for immune and inflammatory responses in tissues. However, it is unclear if Ca2+ /CaM signalling interferes with muscle inflammation. Here we investigated the roles of CaM-dependent signalling in muscle inflammation in mice that had acute myoinjuries in the tibialis anterior muscle induced by intramuscular cardiotoxin (CTX) injections and received intraperitoneal injections of either the CaM inhibitor calmidazolium chloride (CCL) or CaM agonist calcium-like peptide 1 (CALP1). Multiple inflammatory parameters, including muscle autoantigens and toll-like receptors, mononuclear cell infiltration, cytokines and chemokines associated with peripheral muscle inflammation, were examined after the injury and treatment. CALP1 treatment enhanced intramuscular infiltration of monocytes/macrophages into the damaged tibialis anterior muscle and up-regulated mRNA and protein levels of muscle autoantigens (Mi-2, HARS and Ku70) and Toll-like receptor 3 (TLR3), and mRNA levels of tumor necrosis factor α (TNF-α), interleukin-6 (IL-6), Monocyte chemoattractant protein-1 (MCP1), Monocyte chemoattractant protein-3 (MCP3) and Macrophage inflammatory protein-1(MIP-1α) in damaged muscle. In contrast, CCL treatment decreased the intramuscular cell infiltration and mRNA levels of the inflammatory mediators. After CALP1 treatment, a substantial up-regulation in Ca2+ /calmodulin-dependent protein kinase II (CaMKII), Ca2+ /calmodulin-dependent protein kinase IV (CaMKIV) and nuclear factor of activated T cells (NFAT) activity was detected in CD45+ cells isolated from the damaged muscle. More pro-inflammatory F4/80+ Ly-6C+ cells were detected in CD45-gated cells after CALP1 treatment than in those after CCL treatment or no treatment. Consistently, in interferon-γ-stimulated cultured myoblasts and myotubes, CALP1 treatment up-regulated the activities of CaMKII, CaMKIV and NFAT, and levels of class I/II major histocompatibility complexes (MHC-I/II) and TLR3. Our findings demonstrated that CaM-dependent signalling pathways mediate the injury-induced acute muscle inflammatory response.
Collapse
Affiliation(s)
- Jijie Hu
- Department of Orthopaedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Dandan Shi
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Hubei Key Laboratory of Embryonic Stem Cell Research, Department of Anatomy, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, China
| | - Maochao Ding
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Department of Human Anatomy, Tissue Repair and Regenerative Medicine Research Center, Wenzhou Medical University, Wenzhou, 325035, China
| | - Tao Huang
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ruicai Gu
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jiangwei Xiao
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Cory J Xian
- School of Pharmacy and Medical Sciences, and UniSA Cancer Research Institute, University of South Australia, Adelaide, Australia
| | - Jianghui Dong
- School of Pharmacy and Medical Sciences, and UniSA Cancer Research Institute, University of South Australia, Adelaide, Australia
| | - Liping Wang
- School of Pharmacy and Medical Sciences, and UniSA Cancer Research Institute, University of South Australia, Adelaide, Australia
| | - Hua Liao
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
48
|
Gang W, Yu-Zhu W, Yang Y, Feng S, Xing-Li F, Heng Z. The critical role of calcineurin/NFAT (C/N) pathways and effective antitumor prospect for colorectal cancers. J Cell Biochem 2019; 120:19254-19273. [PMID: 31489709 DOI: 10.1002/jcb.29243] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 06/10/2019] [Indexed: 12/11/2022]
Abstract
Transcription factors (TFs) like a nuclear factor of activated T-cells (NFAT) and its controller calcineurin are highly expressed in primary intestinal epithelial cells (IECs) due to delamination, damage by tumor-associated flora and selective activation in the intestinal tract tumor are crucial in the progression and growth of colorectal cancer (CRC). This study sought to summarize the current findings concerning the dysregulated calcineurin/NFAT (C/N) signaling involved in CRC initiation and progression. These signalings include proliferation, T-cell functions, and glycolysis with high lactate production that remodels the acidosis, which genes in tumor cells provide an evolutionary advantage, or even increased their attack phenotype. Moreover, the relationship between C/N and gut microbiome in CRC, especially role of NFAT and toll-like receptor signaling in regulating intestinal microbiota are also discussed. Furthermore, this review will discuss the proteins and genes relating to C/N induced acidosis in CRC, which includes ASIC2 regulated C/N1 and TFs associated with the glycolytic by-product that affect T-cell functions and CRC cell growth. It is revealed that calcineurin or NFAT targeting to antitumor, selective calcineurin inhibition or targets in NFAT signaling may be useful for clinical treatment of CRC. This can further aid in the identification of specific targets via cancer patient-personalized approach. Future studies should be focused on targeting to C/N or TLR signaling by the combination of therapeutic agents to regulate T-cell functions and gut microbiome for activating potent anticancer property with the prospect of potentiating the antitumor therapy for CRC.
Collapse
Affiliation(s)
- Wang Gang
- Department of Pharmaceutics, Shanghai Eight People's Hospital, Jiangsu University, Shanghai, China
| | - Wang Yu-Zhu
- Department of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yu Yang
- Department of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Shi Feng
- Department of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Fu Xing-Li
- Department of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Zhang Heng
- Department of General Surgery, Nanjing Lishui District People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, China
| |
Collapse
|
49
|
Wang T, Xu Y, Yuan Y, Xu P, Zhang C, Li F, Wang L, Yin C, Zhang L, Cai X, Zhu C, Xu J, Liang B, Schaul S, Xie P, Yue D, Liao Z, Yu L, Luo L, Zhou G, Yang J, He Z, Du M, Zhou Y, Deng B, Wang S, Gao P, Zhu X, Xi Q, Zhang Y, Shu G, Jiang Q. Succinate induces skeletal muscle fiber remodeling via SUNCR1 signaling. EMBO Rep 2019; 20:e47892. [PMID: 31318145 PMCID: PMC6727026 DOI: 10.15252/embr.201947892] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 06/13/2019] [Accepted: 06/26/2019] [Indexed: 01/08/2023] Open
Abstract
The conversion of skeletal muscle fiber from fast twitch to slow-twitch is important for sustained and tonic contractile events, maintenance of energy homeostasis, and the alleviation of fatigue. Skeletal muscle remodeling is effectively induced by endurance or aerobic exercise, which also generates several tricarboxylic acid (TCA) cycle intermediates, including succinate. However, whether succinate regulates muscle fiber-type transitions remains unclear. Here, we found that dietary succinate supplementation increased endurance exercise ability, myosin heavy chain I expression, aerobic enzyme activity, oxygen consumption, and mitochondrial biogenesis in mouse skeletal muscle. By contrast, succinate decreased lactate dehydrogenase activity, lactate production, and myosin heavy chain IIb expression. Further, by using pharmacological or genetic loss-of-function models generated by phospholipase Cβ antagonists, SUNCR1 global knockout, or SUNCR1 gastrocnemius-specific knockdown, we found that the effects of succinate on skeletal muscle fiber-type remodeling are mediated by SUNCR1 and its downstream calcium/NFAT signaling pathway. In summary, our results demonstrate succinate induces transition of skeletal muscle fiber via SUNCR1 signaling pathway. These findings suggest the potential beneficial use of succinate-based compounds in both athletic and sedentary populations.
Collapse
Affiliation(s)
- Tao Wang
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Ya‐Qiong Xu
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Ye‐Xian Yuan
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Ping‐Wen Xu
- Division of EndocrinologyDepartment of MedicineThe University of Illinois at ChicagoChicagoILUSA
| | - Cha Zhang
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Fan Li
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Li‐Na Wang
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Cong Yin
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Lin Zhang
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Xing‐Cai Cai
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Can‐Jun Zhu
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Jing‐Ren Xu
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Bing‐Qing Liang
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Sarah Schaul
- Division of EndocrinologyDepartment of MedicineThe University of Illinois at ChicagoChicagoILUSA
| | - Pei‐Pei Xie
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Dong Yue
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Zheng‐Rui Liao
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Lu‐Lu Yu
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Lv Luo
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Gan Zhou
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Jin‐Ping Yang
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Zhi‐Hui He
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Man Du
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Yu‐Ping Zhou
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Bai‐Chuan Deng
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Song‐Bo Wang
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Ping Gao
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Xiao‐Tong Zhu
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Qian‐Yun Xi
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Yong‐Liang Zhang
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Gang Shu
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
- National Engineering Research Center for Breeding Swine IndustryCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Qing‐Yan Jiang
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
- National Engineering Research Center for Breeding Swine IndustryCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| |
Collapse
|
50
|
Wang L, Zhu Y, Liu X, Chao Z, Wang Y, Zhong T, Guo J, Zhan S, Li L, Zhang H. Glycogen synthase kinase 3β (GSK3β) regulates the expression of MyHC2a in goat skeletal muscle satellite cells (SMSCs). Anim Sci J 2019; 90:1042-1049. [PMID: 31237073 DOI: 10.1111/asj.13253] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 05/12/2019] [Accepted: 05/27/2019] [Indexed: 12/16/2022]
Abstract
Glycogen synthase kinase beta (GSK3β) plays an important role in skeletal muscle growth, regeneration, and repair. However, the mechanism of GSK3β regulating MyHC2a expression is currently not clear. In this study, GSK3β inhibition promoted skeletal muscle satellite cells (SMSCs) differentiation and increased expression of MyoD, MyoG, MyHC1, and MyHC2a genes. Then we cloned approximately 1.1 kb of goat MyHC2a gene promoter. The deletion fragment (-514/+55) of MyHC2a promoter exhibited the highest level of promoter activity, and a NFATc2 element in this region was responsible for MyHC2a promoter activity. Treatment of SB216713 significantly decreased the transcriptional activity of the fragment (-514/+55). Furthermore, GSK3β inhibition had no effect on the luciferase activity of MyHC2a promoter after mutating the NFATc2-binding site. These results demonstrated that GSK3β inhibition promoted SMSCs differentiation and regulated the MyHC2a gene expression through NFATc2 in goat-differentiated SMSCs.
Collapse
Affiliation(s)
- Linjie Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, P.R. China
| | - Yuehua Zhu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, P.R. China
| | - Xin Liu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, P.R. China
| | - Zhe Chao
- Institute of Animal Science and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, P.R. China
| | - Yan Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, P.R. China
| | - Tao Zhong
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, P.R. China
| | - Jiazhong Guo
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, P.R. China
| | - Siyuan Zhan
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, P.R. China
| | - Li Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, P.R. China
| | - Hongping Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, P.R. China
| |
Collapse
|