1
|
Onnée M, Malfatti E. The widening genetic and myopathologic spectrum of congenital myopathies (CMYOs): a narrative review. Neuromuscul Disord 2025; 49:105338. [PMID: 40112751 DOI: 10.1016/j.nmd.2025.105338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/28/2025] [Accepted: 03/04/2025] [Indexed: 03/22/2025]
Abstract
Congenital myopathies (CMYOs) represent a genetically and clinically heterogeneous group of disorders characterized by early-onset muscle weakness and distinct myopathologic features. The advent of next-generation sequencing (NGS) has accelerated the identification of causative genes, leading to the discovery of novel CMYOs and thereby challenging the traditional classification. In this comprehensive review, we focus on the clinical, myopathologic, molecular and pathophysiological features of 33 newly identified CMYOs.
Collapse
Affiliation(s)
- Marion Onnée
- Institut Mondor de Recherche Biomédicale, Université Paris Est Créteil, Institut National de la Santé et de la Recherche Médicale U955, 94010 Créteil, France
| | - Edoardo Malfatti
- Institut Mondor de Recherche Biomédicale, Université Paris Est Créteil, Institut National de la Santé et de la Recherche Médicale U955, 94010 Créteil, France; Assistance Publique-Hôpitaux de Paris, Centre de Référence de Pathologie Neuromusculaire Nord-Est-Ile-de-France, Filnemus, Hôpital Henri Mondor, 94010 Créteil, France; European Reference Center for Neuromuscular Disorders, EURO-NMD, France.
| |
Collapse
|
2
|
Battey E, Dos Santos M, Biswas D, Maire P, Sakamoto K. Protocol for muscle fiber type and cross-sectional area analysis in cryosections of whole lower mouse hindlimbs. STAR Protoc 2024; 5:103424. [PMID: 39487985 PMCID: PMC11565383 DOI: 10.1016/j.xpro.2024.103424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/01/2024] [Accepted: 10/10/2024] [Indexed: 11/04/2024] Open
Abstract
We outline a protocol to visualize all mouse lower hindlimb skeletal muscles simultaneously. We describe procedures for orientating the whole lower hindlimb in gum tragacanth prior to freezing, simplifying the proceeding experimental steps, and enhancing the comprehensiveness of characterizations. We then detail steps for quantifying muscle fiber size and fiber type characteristics in a single cryosection using histochemistry and immunofluorescence. This protocol can be applied to histological and (immuno)histochemical evaluations such as muscle regeneration, fibrosis, enzymatic activity, and glycogen content.
Collapse
Affiliation(s)
- Edmund Battey
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Capital Region, Denmark; Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Capital Region, Denmark.
| | - Matthieu Dos Santos
- Department of Molecular Biology, the Hamon Center for Regenerative Science and Medicine, and Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Dipsikha Biswas
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Capital Region, Denmark
| | - Pascal Maire
- Université Paris Cité, Institut Cochin, INSERM, CNRS, 75014 Paris, France
| | - Kei Sakamoto
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Capital Region, Denmark.
| |
Collapse
|
3
|
Poliacikova G, Aouane A, Caruso N, Brouilly N, Maurel-Zaffran C, Graba Y, Saurin AJ. The Hox protein Antennapedia orchestrates Drosophila adult flight muscle development. SCIENCE ADVANCES 2024; 10:eadr2261. [PMID: 39602537 PMCID: PMC11601212 DOI: 10.1126/sciadv.adr2261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024]
Abstract
Muscle development and diversity require a large number of spatially and temporally regulated events controlled by transcription factors (TFs). Drosophila has long stood as a model to study myogenesis due to the highly conserved key TFs involved at all stages of muscle development. While many studies focused on the diversification of Drosophila larval musculature, how distinct adult muscle types are generated is much less characterized. Here, we identify an essential regulator of Drosophila thoracic flight muscle development, the Hox TF Antennapedia (Antp). Correcting a long-standing belief that flight muscle development occurs without the input of Hox TFs, we show that Antp intervenes at several stages of flight muscle development, from the establishment of the progenitor pool in the embryo to myoblast differentiation in the early pupa. Furthermore, the precisely regulated clearance of Hox in the developing flight muscle fibers is required to allow for fibrillar muscle fate diversification, setting these muscles apart from all other adult tubular muscle types.
Collapse
Affiliation(s)
- Gabriela Poliacikova
- Aix-Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), UMR 7288, Case 907, Parc Scientifique de Luminy, Marseille Cedex 09 13288, France
| | - Aïcha Aouane
- Aix-Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), UMR 7288, Case 907, Parc Scientifique de Luminy, Marseille Cedex 09 13288, France
| | - Nathalie Caruso
- Aix-Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), UMR 7288, Case 907, Parc Scientifique de Luminy, Marseille Cedex 09 13288, France
| | - Nicolas Brouilly
- Aix-Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), UMR 7288, Case 907, Parc Scientifique de Luminy, Marseille Cedex 09 13288, France
| | - Corinne Maurel-Zaffran
- Aix-Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), UMR 7288, Case 907, Parc Scientifique de Luminy, Marseille Cedex 09 13288, France
| | - Yacine Graba
- Aix-Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), UMR 7288, Case 907, Parc Scientifique de Luminy, Marseille Cedex 09 13288, France
| | - Andrew J. Saurin
- Aix-Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), UMR 7288, Case 907, Parc Scientifique de Luminy, Marseille Cedex 09 13288, France
| |
Collapse
|
4
|
Jia R, Huang X, Yang J, Wang L, Li J, Li Y, Gun S, Yan Z, Wang P, Yang Q. Gender-Specific DNA Methylation Profiles Associated with Adult Weight in Hezuo Pigs. Int J Mol Sci 2024; 25:11488. [PMID: 39519040 PMCID: PMC11547036 DOI: 10.3390/ijms252111488] [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: 09/07/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
The Hezuo pig, an important native Tibetan breed in China, exhibits differences in adult body weight, with females typically heavier than males. The underlying mechanisms for this disparity remain unclear. DNA methylation changes are known to influence animal growth and development and regulate Hezuo pig growth by altering gene expression related to these processes, thus differentially affecting adult body weight between genders. This study conducted DNA methylation analysis and expression profiling using pituitary tissues from male and female Hezuo pigs at 3 and 8 months old (M3M, M3F, M8M, and M8F). In total 346, 795, 371, and 839 differentially methylated genes (DMGs) were identified in the M3M vs. M3F, M3F vs. M8F, M3M vs. M8M, and M8M vs. M8F groups, respectively. The comparative analysis of differentially methylated regions (DMRs) genes and DEGs (differentially expressed regions) revealed that key genes involved in growth, hormone secretion, and the hypothalamic-pituitary-gonadal axis are primarily enriched in signaling pathways such as PI3K-Akt, Hippo, and adrenergic. Further analysis combining methylation and transcriptomics identified five candidate methylated genes (CCL2, MYL2, GST, CTSH, and MCH) linked to adult body weight in Hezuo pigs. Additionally, the correlation analysis suggested that these genes influence growth and development in boars and sows by regulating the secretion and synthesis of related hormones, leading to heavier weights in females. In conclusion, variations in adult body weight between male and female pigs may stem from the impact of DNA methylation on gene expression related to growth and development. These findings offer new insights into the regulatory mechanisms of DNA methylation during weight gain in Hezuo pigs.
Collapse
Affiliation(s)
- Rui Jia
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (R.J.); (X.H.); (J.Y.); (L.W.); (J.L.); (Y.L.); (S.G.); (Z.Y.); (P.W.)
| | - Xiaoyu Huang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (R.J.); (X.H.); (J.Y.); (L.W.); (J.L.); (Y.L.); (S.G.); (Z.Y.); (P.W.)
| | - Jiaojiao Yang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (R.J.); (X.H.); (J.Y.); (L.W.); (J.L.); (Y.L.); (S.G.); (Z.Y.); (P.W.)
- Gansu Innovations Center for Swine Production Engineering and Technology, Lanzhou 730070, China
| | - Longlong Wang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (R.J.); (X.H.); (J.Y.); (L.W.); (J.L.); (Y.L.); (S.G.); (Z.Y.); (P.W.)
| | - Jie Li
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (R.J.); (X.H.); (J.Y.); (L.W.); (J.L.); (Y.L.); (S.G.); (Z.Y.); (P.W.)
| | - Yao Li
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (R.J.); (X.H.); (J.Y.); (L.W.); (J.L.); (Y.L.); (S.G.); (Z.Y.); (P.W.)
| | - Shuangbao Gun
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (R.J.); (X.H.); (J.Y.); (L.W.); (J.L.); (Y.L.); (S.G.); (Z.Y.); (P.W.)
- Gansu Innovations Center for Swine Production Engineering and Technology, Lanzhou 730070, China
| | - Zunqiang Yan
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (R.J.); (X.H.); (J.Y.); (L.W.); (J.L.); (Y.L.); (S.G.); (Z.Y.); (P.W.)
| | - Pengfei Wang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (R.J.); (X.H.); (J.Y.); (L.W.); (J.L.); (Y.L.); (S.G.); (Z.Y.); (P.W.)
| | - Qiaoli Yang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (R.J.); (X.H.); (J.Y.); (L.W.); (J.L.); (Y.L.); (S.G.); (Z.Y.); (P.W.)
- Gansu Innovations Center for Swine Production Engineering and Technology, Lanzhou 730070, China
| |
Collapse
|
5
|
Wang L, Bu T, Wu X, Li L, Sun F, Cheng CY. Motor proteins, spermatogenesis and testis function. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 141:381-445. [PMID: 38960481 DOI: 10.1016/bs.apcsb.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
The role of motor proteins in supporting intracellular transports of vesicles and organelles in mammalian cells has been known for decades. On the other hand, the function of motor proteins that support spermatogenesis is also well established since the deletion of motor protein genes leads to subfertility and/or infertility. Furthermore, mutations and genetic variations of motor protein genes affect fertility in men, but also a wide range of developmental defects in humans including multiple organs besides the testis. In this review, we seek to provide a summary of microtubule and actin-dependent motor proteins based on earlier and recent findings in the field. Since these two cytoskeletons are polarized structures, different motor proteins are being used to transport cargoes to different ends of these cytoskeletons. However, their involvement in germ cell transport across the blood-testis barrier (BTB) and the epithelium of the seminiferous tubules remains relatively unknown. It is based on recent findings in the field, we have provided a hypothetical model by which motor proteins are being used to support germ cell transport across the BTB and the seminiferous epithelium during the epithelial cycle of spermatogenesis. In our discussion, we have highlighted the areas of research that deserve attention to bridge the gap of research in relating the function of motor proteins to spermatogenesis.
Collapse
Affiliation(s)
- Lingling Wang
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China; Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Tiao Bu
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Xiaolong Wu
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Linxi Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
| | - Fei Sun
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - C Yan Cheng
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China; Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China.
| |
Collapse
|
6
|
Lee SH, Son Y, Choi KJ, Lee CG, Lee HJ. Distinguish response of low-dose radiation with different dose-rate on gene expression of human coronary artery endothelial cells: a bioinformatic study based on transcriptomic sequencing. Int J Radiat Biol 2024; 100:756-766. [PMID: 38489594 DOI: 10.1080/09553002.2024.2324470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 02/05/2024] [Indexed: 03/17/2024]
Abstract
PURPOSE People are exposed to low-dose radiation in medical diagnosis, occupational, or life circumstances, but the effect of low-dose radiation on human health is still controversial. The biological effects of radiation below 100 mGy are still unproven. In this study, we observed the effects of low-dose radiation (100 mGy) on gene expression in human coronary artery endothelial cells (HCAECs) and its effect on molecular signaling. MATERIALS AND METHODS HCAECs were exposed to 100 mGy ionizing radiation at 6 mGy/h (low-dose-rate) or 288 mGy/h (high-dose-rate). After 72 h, total RNA was extracted from sham or irradiated cells for Quant-Seq 3'mRNA-Seq, and bioinformatic analyses were performed using Metascape. Gene profiling was validated using qPCR. RESULTS Compared to the non-irradiated control group, 100 mGy of ionizing radiation at 6 mGy/h altered the expression of 194 genes involved in signaling pathways related to heart contraction, blood circulation, and cardiac myofibril assembly differentially. However, 100 mGy at 288 mGy/h altered expression of 450 genes involved in cell cycle-related signaling pathways, including cell division, nuclear division, and mitosis differentially. Additionally, gene signatures responding to low-dose radiation, including radiation dose-specific gene profiles (HIST1H2AI, RAVER1, and POTEI) and dose-rate-specific gene profiles (MYL2 for the low-dose-rate and DHRS9 and CA14 for the high-dose-rate) were also identified. CONCLUSIONS We demonstrated that 100 mGy low-dose radiation could alter gene expression and molecular signaling pathways at the low-dose-rate and the high-dose-rate differently. Our findings provide evidence for further research on the potential impact of low-dose radiation on cardiovascular function.
Collapse
Affiliation(s)
- Soo-Ho Lee
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences (KIRAMS), Seoul, Republic of Korea
| | - Yeonghoon Son
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences (KIRAMS), Seoul, Republic of Korea
| | - Kyu Jin Choi
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences (KIRAMS), Seoul, Republic of Korea
| | - Chang Geun Lee
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, Republic of Korea
| | - Hae-June Lee
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences (KIRAMS), Seoul, Republic of Korea
| |
Collapse
|
7
|
Sánchez-Baizán N, Jarne-Sanz I, Roco ÁS, Schartl M, Piferrer F. Extraordinary variability in gene activation and repression programs during gonadal sex differentiation across vertebrates. Front Cell Dev Biol 2024; 12:1328365. [PMID: 38322165 PMCID: PMC10844511 DOI: 10.3389/fcell.2024.1328365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 01/11/2024] [Indexed: 02/08/2024] Open
Abstract
Genes involved in gonadal sex differentiation have been traditionally thought to be fairly conserved across vertebrates, but this has been lately questioned. Here, we performed the first comparative analysis of gonadal transcriptomes across vertebrates, from fish to mammals. Our results unambiguously show an extraordinary overall variability in gene activation and repression programs without a phylogenetic pattern. During sex differentiation, genes such as dmrt1, sox9, amh, cyp19a and foxl2 were consistently either male- or female-enriched across species while many genes with the greatest expression change within each sex were not. We also found that downregulation in the opposite sex, which had only been quantified in the mouse model, was also prominent in the rest of vertebrates. Finally, we report 16 novel conserved markers (e.g., fshr and dazl) and 11 signaling pathways. We propose viewing vertebrate gonadal sex differentiation as a hierarchical network, with conserved hub genes such as sox9 and amh alongside less connected and less conserved nodes. This proposed framework implies that evolutionary pressures may impact genes based on their level of connectivity.
Collapse
Affiliation(s)
- Núria Sánchez-Baizán
- Institut de Ciències del Mar (ICM), Spanish National Research Council (CSIC), Barcelona, Spain
| | - Ignasi Jarne-Sanz
- Institut de Ciències del Mar (ICM), Spanish National Research Council (CSIC), Barcelona, Spain
| | - Álvaro S. Roco
- Developmental Biochemistry, Biocenter, University of Wuerzburg, Wuerzburg, Germany
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaén, Jaén, Spain
| | - Manfred Schartl
- Developmental Biochemistry, Biocenter, University of Wuerzburg, Wuerzburg, Germany
- Xiphophorus Genetic Stock Center, Texas State University, San Marcos, TX, United States
| | - Francesc Piferrer
- Institut de Ciències del Mar (ICM), Spanish National Research Council (CSIC), Barcelona, Spain
| |
Collapse
|
8
|
Cao CR, Seidman MA. Multiverse of Mutations: Novel Phenotype-Genotype Correlation. Can J Cardiol 2023; 39:1335-1337. [PMID: 37348758 DOI: 10.1016/j.cjca.2023.06.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 06/24/2023] Open
Affiliation(s)
- Cathy R Cao
- Laboratory Medicine Program, University Health Network, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Michael A Seidman
- Laboratory Medicine Program, University Health Network, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
9
|
Borowczyk M, Dobosz P, Szczepanek-Parulska E, Budny B, Dębicki S, Filipowicz D, Wrotkowska E, Oszywa M, Verburg FA, Janicka-Jedyńska M, Ziemnicka K, Ruchała M. Follicular Thyroid Adenoma and Follicular Thyroid Carcinoma-A Common or Distinct Background? Loss of Heterozygosity in Comprehensive Microarray Study. Cancers (Basel) 2023; 15:638. [PMID: 36765597 PMCID: PMC9913827 DOI: 10.3390/cancers15030638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 01/22/2023] Open
Abstract
Pre- and postsurgical differentiation between follicular thyroid adenoma (FTA) and follicular thyroid cancer (FTC) represents a significant diagnostic challenge. Furthermore, it remains unclear whether they share a common or distinct background and what the mechanisms underlying follicular thyroid lesions malignancy are. The study aimed to compare FTA and FTC by the comprehensive microarray and to identify recurrent regions of loss of heterozygosity (LOH). We analyzed formalin-fixed paraffin-embedded (FFPE) samples acquired from 32 Caucasian patients diagnosed with FTA (16) and FTC (16). We used the OncoScan™ microarray assay (Affymetrix, USA), using highly multiplexed molecular inversion probes for single nucleotide polymorphism (SNP). The total number of LOH was higher in FTC compared with FTA (18 vs. 15). The most common LOH present in 21 cases, in both FTA (10 cases) and FTC (11 cases), was 16p12.1, which encompasses many cancer-related genes, such as TP53, and was followed by 3p21.31. The only LOH present exclusively in FTA patients (56% vs. 0%) was 11p11.2-p11.12. The alteration which tended to be detected more often in FTC (6 vs. 1 in FTA) was 12q24.11-q24.13 overlapping FOXN4, MYL2, PTPN11 genes. FTA and FTC may share a common genetic background, even though differentiating rearrangements may also be detected.
Collapse
Affiliation(s)
- Martyna Borowczyk
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland
- Department of Medical Simulation, Poznan University of Medical Sciences, 60-806 Poznan, Poland
| | - Paula Dobosz
- Department of Genetics and Genomics, Central Clinical Hospital of the Ministry of Interior Affairs and Administration, 02-507 Warsaw, Poland
| | - Ewelina Szczepanek-Parulska
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Bartłomiej Budny
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Szymon Dębicki
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Dorota Filipowicz
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Elżbieta Wrotkowska
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Michalina Oszywa
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Frederik A. Verburg
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
| | | | - Katarzyna Ziemnicka
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Marek Ruchała
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| |
Collapse
|
10
|
Zhang S, Zhang Y, Chen C, Hu Q, Fu Y, Xu L, Wang C, Liu Y. Identification of Robust and Key Differentially Expressed Genes during C2C12 Cell Myogenesis Based on Multiomics Data. Int J Mol Sci 2022; 23:ijms23116002. [PMID: 35682680 PMCID: PMC9180599 DOI: 10.3390/ijms23116002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/22/2022] [Accepted: 05/24/2022] [Indexed: 02/01/2023] Open
Abstract
Myogenesis is a central step in prenatal myofiber formation, postnatal myofiber hypertrophy, and muscle damage repair in adulthood. RNA-Seq technology has greatly helped reveal the molecular mechanism of myogenesis, but batch effects in different experiments inevitably lead to misinterpretation of differentially expressed genes (DEGs). We previously applied the robust rank aggregation (RRA) method to effectively circumvent batch effects across multiple RNA-Seq datasets from 3T3-L1 cells. Here, we also used the RRA method to integrate nine RNA-Seq datasets from C2C12 cells and obtained 3140 robust DEGs between myoblasts and myotubes, which were then validated with array expression profiles and H3K27ac signals. The upregulated robust DEGs were highly enriched in gene ontology (GO) terms related to muscle cell differentiation and development. Considering that the cooperative binding of transcription factors (TFs) to enhancers to regulate downstream gene expression is a classical epigenetic mechanism, differentially expressed TFs (DETFs) were screened, and potential novel myogenic factors (MAF, BCL6, and ESR1) with high connection degree in protein-protein interaction (PPI) network were presented. Moreover, KLF5 cooperatively binds with the three key myogenic factors (MYOD, MYOG, and MEF2D) in C2C12 cells. Motif analysis speculates that the binding of MYOD and MYOG is KLF5-independent, while MEF2D is KLF5-dependent. It was revealed that KLF5-binding sites could be exploited to filter redundant MYOD-, MYOG-, and MEF2D-binding sites to focus on key enhancers for myogenesis. Further functional annotation of KLF5-binding sites suggested that KLF5 may regulate myogenesis through the PI3K-AKt signaling pathway, Rap1 signaling pathway, and the Hippo signaling pathway. In general, our study provides a wealth of untapped candidate targets for myogenesis and contributes new insights into the core regulatory mechanisms of myogenesis relying on KLF5-binding signal.
Collapse
Affiliation(s)
- Song Zhang
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China; (S.Z.); (Y.Z.); (C.C.); (Q.H.); (Y.F.); (L.X.); (C.W.)
- Innovation Group of Pig Genome Design and Breeding, Research Centre for Animal Genome, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
| | - Yuanyuan Zhang
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China; (S.Z.); (Y.Z.); (C.C.); (Q.H.); (Y.F.); (L.X.); (C.W.)
- Innovation Group of Pig Genome Design and Breeding, Research Centre for Animal Genome, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
- School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Choulin Chen
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China; (S.Z.); (Y.Z.); (C.C.); (Q.H.); (Y.F.); (L.X.); (C.W.)
- Innovation Group of Pig Genome Design and Breeding, Research Centre for Animal Genome, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Qingqing Hu
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China; (S.Z.); (Y.Z.); (C.C.); (Q.H.); (Y.F.); (L.X.); (C.W.)
- Innovation Group of Pig Genome Design and Breeding, Research Centre for Animal Genome, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Yang Fu
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China; (S.Z.); (Y.Z.); (C.C.); (Q.H.); (Y.F.); (L.X.); (C.W.)
- Innovation Group of Pig Genome Design and Breeding, Research Centre for Animal Genome, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
| | - Lingna Xu
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China; (S.Z.); (Y.Z.); (C.C.); (Q.H.); (Y.F.); (L.X.); (C.W.)
- Innovation Group of Pig Genome Design and Breeding, Research Centre for Animal Genome, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
| | - Chao Wang
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China; (S.Z.); (Y.Z.); (C.C.); (Q.H.); (Y.F.); (L.X.); (C.W.)
- Innovation Group of Pig Genome Design and Breeding, Research Centre for Animal Genome, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Yuwen Liu
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China; (S.Z.); (Y.Z.); (C.C.); (Q.H.); (Y.F.); (L.X.); (C.W.)
- Innovation Group of Pig Genome Design and Breeding, Research Centre for Animal Genome, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
- Kunpeng Institute of Modern Agriculture at Foshan, Chinese Academy of Agricultural Sciences, Foshan 528226, China
- Correspondence:
| |
Collapse
|
11
|
Genetic Insights into Primary Restrictive Cardiomyopathy. J Clin Med 2022; 11:jcm11082094. [PMID: 35456187 PMCID: PMC9027761 DOI: 10.3390/jcm11082094] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 12/04/2022] Open
Abstract
Restrictive cardiomyopathy is a rare cardiac disease causing severe diastolic dysfunction, ventricular stiffness and dilated atria. In consequence, it induces heart failure often with preserved ejection fraction and is associated with a high mortality. Since it is a poor clinical prognosis, patients with restrictive cardiomyopathy frequently require heart transplantation. Genetic as well as non-genetic factors contribute to restrictive cardiomyopathy and a significant portion of cases are of unknown etiology. However, the genetic forms of restrictive cardiomyopathy and the involved molecular pathomechanisms are only partially understood. In this review, we summarize the current knowledge about primary genetic restrictive cardiomyopathy and describe its genetic landscape, which might be of interest for geneticists as well as for cardiologists.
Collapse
|
12
|
Bang ML, Bogomolovas J, Chen J. Understanding the molecular basis of cardiomyopathy. Am J Physiol Heart Circ Physiol 2022; 322:H181-H233. [PMID: 34797172 PMCID: PMC8759964 DOI: 10.1152/ajpheart.00562.2021] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/16/2021] [Accepted: 11/16/2021] [Indexed: 02/03/2023]
Abstract
Inherited cardiomyopathies are a major cause of mortality and morbidity worldwide and can be caused by mutations in a wide range of proteins located in different cellular compartments. The present review is based on Dr. Ju Chen's 2021 Robert M. Berne Distinguished Lectureship of the American Physiological Society Cardiovascular Section, in which he provided an overview of the current knowledge on the cardiomyopathy-associated proteins that have been studied in his laboratory. The review provides a general summary of the proteins in different compartments of cardiomyocytes associated with cardiomyopathies, with specific focus on the proteins that have been studied in Dr. Chen's laboratory.
Collapse
Affiliation(s)
- Marie-Louise Bang
- Institute of Genetic and Biomedical Research (IRGB), National Research Council (CNR), Milan Unit, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Julius Bogomolovas
- Division of Cardiovascular Medicine, Department of Medicine Cardiology, University of California, San Diego, La Jolla, California
| | - Ju Chen
- Division of Cardiovascular Medicine, Department of Medicine Cardiology, University of California, San Diego, La Jolla, California
| |
Collapse
|
13
|
Differential gene expression by RNA-seq during Alzheimer’s disease-like progression in the Drosophila melanogaster model. Neurosci Res 2022; 180:1-12. [DOI: 10.1016/j.neures.2022.02.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 02/16/2022] [Accepted: 02/20/2022] [Indexed: 01/10/2023]
|
14
|
Mak RH, Querfeld U, Gonzalez A, Gunta S, Cheung WW. Differential Effects of 25-Hydroxyvitamin D 3 versus 1α 25-Dihydroxyvitamin D 3 on Adipose Tissue Browning in CKD-Associated Cachexia. Cells 2021; 10:3382. [PMID: 34943890 PMCID: PMC8699879 DOI: 10.3390/cells10123382] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/22/2021] [Accepted: 11/26/2021] [Indexed: 11/16/2022] Open
Abstract
Patients with chronic kidney disease (CKD) often have low serum concentrations of 25(OH)D3 and 1,25(OH)2D3. We investigated the differential effects of 25(OH)D3 versus 1,25(OH)2D3 repletion in mice with surgically induced CKD. Intraperitoneal supplementation of 25(OH)D3 (75 μg/kg/day) or 1,25(OH)2D3 (60 ng/kg/day) for 6 weeks normalized serum 25(OH)D3 or 1,25(OH)2D3 concentrations in CKD mice, respectively. Repletion of 25(OH)D3 normalized appetite, significantly improved weight gain, increased fat and lean mass content and in vivo muscle function, as well as attenuated elevated resting metabolic rate relative to repletion of 1,25(OH)2D3 in CKD mice. Repletion of 25(OH)D3 in CKD mice attenuated adipose tissue browning as well as ameliorated perturbations of energy homeostasis in adipose tissue and skeletal muscle, whereas repletion of 1,25(OH)2D3 did not. Significant improvement of muscle fiber size and normalization of fat infiltration of gastrocnemius was apparent with repletion of 25(OH)D3 but not with 1,25(OH)2D3 in CKD mice. This was accompanied by attenuation of the aberrant gene expression of muscle mass regulatory signaling, molecular pathways related to muscle fibrosis as well as muscle expression profile associated with skeletal muscle wasting in CKD mice. Our findings provide evidence that repletion of 25(OH)D3 exerts metabolic advantages over repletion of 1,25(OH)2D3 by attenuating adipose tissue browning and muscle wasting in CKD mice.
Collapse
Affiliation(s)
- Robert H. Mak
- Division of Pediatric Nephrology, Rady Children’s Hospital, University of California, San Diego, CA 92093, USA; (A.G.); (S.G.); (W.W.C.)
| | - Uwe Querfeld
- Department of Paediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany;
| | - Alex Gonzalez
- Division of Pediatric Nephrology, Rady Children’s Hospital, University of California, San Diego, CA 92093, USA; (A.G.); (S.G.); (W.W.C.)
| | - Sujana Gunta
- Division of Pediatric Nephrology, Rady Children’s Hospital, University of California, San Diego, CA 92093, USA; (A.G.); (S.G.); (W.W.C.)
- Pediatric Services, Vista Community Clinic, Vista, CA 92084, USA
| | - Wai W. Cheung
- Division of Pediatric Nephrology, Rady Children’s Hospital, University of California, San Diego, CA 92093, USA; (A.G.); (S.G.); (W.W.C.)
| |
Collapse
|
15
|
Wang J, Gao S, Dong K, Guo P, Shan MJ. MYL2 as a potential predictive biomarker for rhabdomyosarcoma. Medicine (Baltimore) 2021; 100:e27101. [PMID: 34596111 PMCID: PMC8483830 DOI: 10.1097/md.0000000000027101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 08/12/2021] [Indexed: 01/05/2023] Open
Abstract
Rhabdomyosarcoma (RMS) is a common malignant soft tissue sarcoma, which is the third most common soft tissue sarcoma after malignant fibrohistoma and liposarcoma. The discovery of potential postbiomarkers could lead to early and more effective treatment measures to reduce the mortality of RMS. The discovery of biomarker is expected to be the direction of targeted therapy, providing a new direction for the precise treatment of RMS.Gene Expression Omnibus database was used to download the tow gene profiles, GSE28511 and GSE135517. GEO2R was applied to identify differently expressed genes (DEGs) between RMS and normal group. Database for Annotation, Visualization and Integrated Discovery and Metascape can perform the enrichment analysis for the DEGs. Protein-protein interaction network was constructed, and the hub genes was identified by the Cytoscape. Expression and overall survival analysis of hub genes were performed.A total of 15 common DEGs were screened between RMS and normal tissues. The enrichment analysis here showed that the DEGs mainly enriched in the muscle filament sliding, myofibril, protein complex, sarcomere, myosin complex, nuclear chromosome, and tight junction. The 6 hub genes (DNA Topoisomerase II Alpha, Insulin Like Growth Factor 2, HIST1H4C, Cardiomyopathy Associated 5, Myosin Light Chain 2 [MYL2], Myosin Heavy Chain 2) were identified. Compared with the normal tissues, MYL2 were down-regulated in the RMS tissues. RMS patients with low expression level of MYL2 had poorer overall survival times than those with high expression levels (P < .05).In summary, lower expression of MYL2 was 1 prediction for poor prognosis of RMS. MYL2 is hope to be the target of therapy, which leads to more effective treatment and reduces the mortality rate of RMS.
Collapse
Affiliation(s)
- Junning Wang
- The second Department of orthopedics, Hangzhou Fuyang District First People's Hospital, No. 429 Beihuan Road, Fuyang District, Hangzhou 311499, P.R. China
| | - Shang Gao
- Bethune Second Clinical Medical College of Jilin University, 218Ziqiang Hutong, Nanguan District, Changchun City, Jilin Province 130041, China
| | - Keqin Dong
- School of Basic Medical Sciences, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, Hebei 050017, P.R. China
| | - Peiyuan Guo
- School of Basic Medical Sciences, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, Hebei 050017, P.R. China
| | - Meng-jie Shan
- Graduate School, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 9 Dongdansantiao, Dongcheng District, Beijing 100730, China
- Department of plastic surgery, Peking Union Medical College Hospital, Beijing, 100730, China
| |
Collapse
|
16
|
Gonzalez A, Cheung WW, Perens EA, Oliveira EA, Gertler A, Mak RH. A Leptin Receptor Antagonist Attenuates Adipose Tissue Browning and Muscle Wasting in Infantile Nephropathic Cystinosis-Associated Cachexia. Cells 2021; 10:1954. [PMID: 34440723 PMCID: PMC8393983 DOI: 10.3390/cells10081954] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/19/2021] [Accepted: 07/24/2021] [Indexed: 12/12/2022] Open
Abstract
Mice lacking the functional cystinosin gene (Ctns-/-), a model of infantile nephropathic cystinosis (INC), exhibit the cachexia phenotype with adipose tissue browning and muscle wasting. Elevated leptin signaling is an important cause of chronic kidney disease-associated cachexia. The pegylated leptin receptor antagonist (PLA) binds to but does not activate the leptin receptor. We tested the efficacy of this PLA in Ctns-/- mice. We treated 12-month-old Ctns-/- mice and control mice with PLA (7 mg/kg/day, IP) or saline as a vehicle for 28 days. PLA normalized food intake and weight gain, increased fat and lean mass, decreased metabolic rate and improved muscle function. It also attenuated perturbations of energy homeostasis in adipose tissue and muscle in Ctns-/- mice. PLA attenuated adipose tissue browning in Ctns-/- mice. PLA increased gastrocnemius weight and fiber size as well as attenuated muscle fat infiltration in Ctns-/- mice. This was accompanied by correcting the increased expression of muscle wasting signaling while promoting the decreased expression of myogenesis in gastrocnemius of Ctns-/- mice. PLA attenuated aberrant expressed muscle genes that have been associated with muscle atrophy, increased energy expenditure and lipolysis in Ctns-/- mice. Leptin antagonism may represent a viable therapeutic strategy for adipose tissue browning and muscle wasting in INC.
Collapse
MESH Headings
- Adipose Tissue, Brown/drug effects
- Adipose Tissue, Brown/metabolism
- Adipose Tissue, Brown/pathology
- Adipose Tissue, White/drug effects
- Adipose Tissue, White/metabolism
- Adipose Tissue, White/pathology
- Amino Acid Transport Systems, Neutral/genetics
- Amino Acid Transport Systems, Neutral/metabolism
- Animals
- Body Composition/drug effects
- Cachexia/etiology
- Cachexia/metabolism
- Cachexia/pathology
- Cachexia/prevention & control
- Cystinosis/complications
- Cystinosis/drug therapy
- Cystinosis/metabolism
- Cystinosis/pathology
- Disease Models, Animal
- Hormone Antagonists/pharmacology
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscular Atrophy/etiology
- Muscular Atrophy/metabolism
- Muscular Atrophy/pathology
- Muscular Atrophy/prevention & control
- Receptors, Leptin/antagonists & inhibitors
- Receptors, Leptin/metabolism
- Signal Transduction
- Mice
Collapse
Affiliation(s)
- Alex Gonzalez
- Division of Pediatric Nephrology, Rady Children’s Hospital, University of California, San Diego, CA 92093-0831, USA; (A.G.); (W.W.C.); (E.A.P.); (E.A.O.)
| | - Wai W. Cheung
- Division of Pediatric Nephrology, Rady Children’s Hospital, University of California, San Diego, CA 92093-0831, USA; (A.G.); (W.W.C.); (E.A.P.); (E.A.O.)
| | - Elliot A. Perens
- Division of Pediatric Nephrology, Rady Children’s Hospital, University of California, San Diego, CA 92093-0831, USA; (A.G.); (W.W.C.); (E.A.P.); (E.A.O.)
| | - Eduardo A. Oliveira
- Division of Pediatric Nephrology, Rady Children’s Hospital, University of California, San Diego, CA 92093-0831, USA; (A.G.); (W.W.C.); (E.A.P.); (E.A.O.)
- Health Sciences Postgraduate Program, School of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte 30130-100, MG, Brazil
| | - Arieh Gertler
- Institute of Biochemistry, Food Science and Nutrition, Hebrew University of Jerusalem, Rehovot 7610001, Israel;
| | - Robert H. Mak
- Division of Pediatric Nephrology, Rady Children’s Hospital, University of California, San Diego, CA 92093-0831, USA; (A.G.); (W.W.C.); (E.A.P.); (E.A.O.)
| |
Collapse
|
17
|
Abstract
PURPOSE OF REVIEW Tremor is a common neurological symptom with a plethora of potential etiologies. Apart from physiological tremor, the vast majority of tremor syndromes are linked to a pacemaker in the central nervous system (CNS) or, less common, in the peripheral nervous system. Myogenic tremor is a novel tremor entity, first reported in 2019 and believed to originate in the muscle itself. In this review, we describe the clinical properties of myogenic tremor and discuss its presumed pathogenesis on the basis of all of the patient cases published so far. RECENT FINDINGS Myogenic tremor manifests itself as a high frequency, postural, and kinetic tremor with onset in infancy. To date, only myopathies affecting the contractile elements, in particular myosin and a myosin-associated protein, have been recognized to feature myogenic tremor. The generator of the tremor is believed to be located in the sarcomere, with propagation and amplification of sarcomeric oscillatory activity through CNS reflex loops, similar to neuropathic tremor. SUMMARY True myogenic tremor must be distinguished from centrally mediated tremor due to myopathies with central nervous system involvement, i.e., mitochondrial myopathies or myotonic dystrophies. The presence of myogenic tremor strongly points toward a sarcomere-associated mutation and may thus be a valuable clinical tool for the differential diagnosis of myopathies.
Collapse
|
18
|
Zhang X, Wang J, Li X, Shen X, Xu D, Tian Y, Huang Y. Transcriptomic investigation of embryonic pectoral muscle reveals increased myogenic processes in Shitou geese compared to Wuzong geese. Br Poult Sci 2021; 62:650-657. [PMID: 33834898 DOI: 10.1080/00071668.2021.1912292] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
1.Embryonic stages before birth are crucial for poultry muscle development, as this determines muscle mass in adulthood. This study characterised the distinction in embryonic pectoral muscle development between Wuzong (WZE, small) and Shitou (STE, large) geese (two indigenous goose breeds in Guangdong Province, China) at embryonic days 15 (E15), 23 (E23) and the day of hatching (P1) to gain insights into the regulatory mechanisms of muscle development.2.The results showed that STE had significantly higher myofibre density during E15-P1 and had significantly larger myofibre diameter at E15 than WZE. By RNA-sequencing analysis, 19 507 genes were detected, and 7121 differentially expressed genes (DEGs) were identified.3.Gene expression distinctions between breeds began increasing from E23, and WZE had different gene expression profiles compared to STE. A GO analysis of DEGs indicated that myo-genes involved at E15 may influence distinct pectoral muscle development characteristics between WZE and STE. The RT-qPCR results were consistent with the RNA-sequencing analysis. Four muscle structure protein coding genes (MYL2, MYL3, TNNI2 and TNNC2 and three other functional genes (CAV3, CACNA1S and NOS1) were identified in a predicted interaction network. These functional genes may interact with muscle structural protein coding genes to regulate embryonic pectoral muscle development in WZE and STE geese.4.The study revealed that STE and WZE had divergent embryonic pectoral muscle development patterns and these differences may begin before E15.
Collapse
Affiliation(s)
- X Zhang
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China.,Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Guangzhou, China
| | - J Wang
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China.,Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Guangzhou, China
| | - X Li
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China.,Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Guangzhou, China
| | - X Shen
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China.,Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Guangzhou, China
| | - D Xu
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China.,Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Guangzhou, China
| | - Y Tian
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China.,Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Guangzhou, China
| | - Y Huang
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China.,Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Guangzhou, China
| |
Collapse
|
19
|
Tamamitsu AM, Nakagama Y, Domoto Y, Yoshida K, Ogawa S, Hirono K, Shindo T, Ogawa Y, Nakano K, Asakai H, Hirata Y, Matsui H, Inuzuka R. Poor Myocardial Compaction in a Patient with Recessive MYL2 Myopathy. Int Heart J 2021; 62:445-447. [PMID: 33731536 DOI: 10.1536/ihj.20-639] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Recessive mutations in the Myosin regulatory light chain 2 (MYL2) gene are the cause of an infantile-onset myopathy, associated with fatal myocardial disease of variable macromorphology. We here present the first Japanese family affected with recessive MYL2 myopathy. Affected siblings manifested typical features and the proband's autopsy findings were compatible with the diagnosis of noncompaction cardiomyopathy. The rapidly progressive clinical course of this recessive MYL2 cardiomyopathy highlights the crucial role of c-terminal tails in MYL2 protein in maintaining cardiac morphology and function.
Collapse
Affiliation(s)
| | - Yu Nakagama
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo.,Department of Parasitology, Graduate School of Medicine, Osaka City University
| | - Yukako Domoto
- Department of Pathology, Graduate School of Medicine, The University of Tokyo
| | - Kenichi Yoshida
- Department of Pathology, Graduate School of Medicine, The University of Tokyo
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University
| | - Keiichi Hirono
- Department of Pediatrics, Graduate School of Medicine, University of Toyama
| | - Takahiro Shindo
- Division of Cardiology, National Center for Child Health and Development
| | - Yosuke Ogawa
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo
| | - Katsutoshi Nakano
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo
| | - Hiroko Asakai
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo
| | - Yoichiro Hirata
- Department of Pediatrics, Kitasato University School of Medicine
| | - Hikoro Matsui
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo
| | - Ryo Inuzuka
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo
| |
Collapse
|
20
|
Sitbon YH, Yadav S, Kazmierczak K, Szczesna-Cordary D. Insights into myosin regulatory and essential light chains: a focus on their roles in cardiac and skeletal muscle function, development and disease. J Muscle Res Cell Motil 2020; 41:313-327. [PMID: 31131433 PMCID: PMC6879809 DOI: 10.1007/s10974-019-09517-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 05/21/2019] [Indexed: 12/15/2022]
Abstract
The activity of cardiac and skeletal muscles depends upon the ATP-coupled actin-myosin interactions to execute the power stroke and muscle contraction. The goal of this review article is to provide insight into the function of myosin II, the molecular motor of the heart and skeletal muscles, with a special focus on the role of myosin II light chain (MLC) components. Specifically, we focus on the involvement of myosin regulatory (RLC) and essential (ELC) light chains in striated muscle development, isoform appearance and their function in normal and diseased muscle. We review the consequences of isoform switching and knockout of specific MLC isoforms on cardiac and skeletal muscle function in various animal models. Finally, we discuss how dysregulation of specific RLC/ELC isoforms can lead to cardiac and skeletal muscle diseases and summarize the effects of most studied mutations leading to cardiac or skeletal myopathies.
Collapse
Affiliation(s)
- Yoel H Sitbon
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, 1600 NW 10th Ave, Miami, FL, 33136, USA
| | - Sunil Yadav
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, 1600 NW 10th Ave, Miami, FL, 33136, USA
| | - Katarzyna Kazmierczak
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, 1600 NW 10th Ave, Miami, FL, 33136, USA
| | - Danuta Szczesna-Cordary
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, 1600 NW 10th Ave, Miami, FL, 33136, USA.
| |
Collapse
|
21
|
Manivannan SN, Darouich S, Masmoudi A, Gordon D, Zender G, Han Z, Fitzgerald-Butt S, White P, McBride KL, Kharrat M, Garg V. Novel frameshift variant in MYL2 reveals molecular differences between dominant and recessive forms of hypertrophic cardiomyopathy. PLoS Genet 2020; 16:e1008639. [PMID: 32453731 PMCID: PMC7274480 DOI: 10.1371/journal.pgen.1008639] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 06/05/2020] [Accepted: 01/29/2020] [Indexed: 12/18/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is characterized by thickening of the ventricular muscle without dilation and is often associated with dominant pathogenic variants in cardiac sarcomeric protein genes. Here, we report a family with two infants diagnosed with infantile-onset HCM and mitral valve dysplasia that led to death before one year of age. Using exome sequencing, we discovered that one of the affected children had a homozygous frameshift variant in Myosin light chain 2 (MYL2:NM_000432.3:c.431_432delCT: p.Pro144Argfs*57;MYL2-fs), which alters the last 20 amino acids of the protein and is predicted to impact the most C-terminal of the three EF-hand domains in MYL2. The parents are unaffected heterozygous carriers of the variant and the variant is absent in control cohorts from gnomAD. The absence of the phenotype in carriers and the infantile presentation of severe HCM is in contrast to HCM associated with dominant MYL2 variants. Immunohistochemical analysis of the ventricular muscle of the deceased patient with the MYL2-fs variant showed a marked reduction of MYL2 expression compared to an unaffected control. In vitro overexpression studies further indicate that the MYL2-fs variant is actively degraded. In contrast, an HCM-associated missense variant (MYL2:p.Gly162Arg) and three other MYL2 stop-gain variants (p.E22*, p.K62*, p.E97*) that result in loss of the EF domains are stably expressed but show impaired localization. The degradation of the MYL2-fs can be rescued by inhibiting the cell’s proteasome function supporting a post-translational effect of the variant. In vivo rescue experiments with a Drosophila MYL2-homolog (Mlc2) knockdown model indicate that neither the MYL2-fs nor the MYL2:p.Gly162Arg variant supports normal cardiac function. The tools that we have generated provide a rapid screening platform for functional assessment of variants of unknown significance in MYL2. Our study supports an autosomal recessive model of inheritance for MYL2 loss-of-function variants in infantile HCM and highlights the variant-specific molecular differences found in MYL2-associated cardiomyopathy. We report a novel frameshift variant in MYL2 that is associated with a severe form of infantile-onset hypertrophic cardiomyopathy. The impact of the variant is only observed in the recessive form of the disease found in the proband and not in the parents who are carriers of the variant. This contrasts with other dominant variants in MYL2 that are associated with cardiomyopathies. We compared the stability of this variant to that of other cardiomyopathy associated MYL2 variants and found molecular differences that correlated with disease pathology. We also show different protein domain requirements for stability and localization of MYL2 in cardiomyocytes. Furthermore, we used a fly model to demonstrate functional deficits due to the variant in the developing heart. Overall, our study shows a molecular mechanism by which loss-of-function variants in MYL2 are recessive while missense variants are dominant. We highlight the use of exome sequencing and functional testing to assist in the diagnosis of rare forms of disease where pathogenicity of the variant is not obvious. The new tools we developed for in vitro functional study and the fly fluorescent reporter analysis will permit rapid analysis of MYL2 variants of unknown significance.
Collapse
Affiliation(s)
- Sathiya N. Manivannan
- Center for Cardiovascular Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, United States of America
- Heart Center, Nationwide Children’s Hospital, Columbus, Ohio, United States of America
| | - Sihem Darouich
- University of Tunis El Manar, Faculty of Medicine of Tunis, LR99ES10 Laboratory of Human Genetics, Tunis, Tunisia
- * E-mail: (SD); (VG)
| | - Aida Masmoudi
- University of Tunis El Manar, Faculty of Medicine of Tunis, Department of Embryo-Fetopathology, Maternity and Neonatology Center, Tunis, Tunisia
| | - David Gordon
- Institute for Genomic Medicine at Nationwide Children’s Hospital, Columbus, Ohio, United States of America
| | - Gloria Zender
- Center for Cardiovascular Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, United States of America
| | - Zhe Han
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Sara Fitzgerald-Butt
- Center for Cardiovascular Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, United States of America
- Heart Center, Nationwide Children’s Hospital, Columbus, Ohio, United States of America
- Department of Pediatrics, The Ohio State University, Columbus, Ohio, United States of America
| | - Peter White
- Institute for Genomic Medicine at Nationwide Children’s Hospital, Columbus, Ohio, United States of America
- Department of Pediatrics, The Ohio State University, Columbus, Ohio, United States of America
| | - Kim L. McBride
- Center for Cardiovascular Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, United States of America
- Heart Center, Nationwide Children’s Hospital, Columbus, Ohio, United States of America
- Department of Pediatrics, The Ohio State University, Columbus, Ohio, United States of America
| | - Maher Kharrat
- University of Tunis El Manar, Faculty of Medicine of Tunis, LR99ES10 Laboratory of Human Genetics, Tunis, Tunisia
| | - Vidu Garg
- Center for Cardiovascular Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, United States of America
- Heart Center, Nationwide Children’s Hospital, Columbus, Ohio, United States of America
- Department of Pediatrics, The Ohio State University, Columbus, Ohio, United States of America
- Department of Molecular Genetics, The Ohio State University, Columbus, Ohio, United States of America
- * E-mail: (SD); (VG)
| |
Collapse
|
22
|
Sarcomeric myopathies associated with tremor: new insights and perspectives. J Muscle Res Cell Motil 2019; 41:285-295. [PMID: 31620961 DOI: 10.1007/s10974-019-09559-1] [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: 05/07/2019] [Accepted: 10/05/2019] [Indexed: 12/21/2022]
Abstract
Myopathies are a large and heterogeneous group of disorders associated with mutations in structural and regulatory genes responsible for proper muscle assembly, organization and function. Despite the molecular diversity of inherited myopathies, they have historically been classified by the phenotypic traits observed in affected patients. It is therefore common for myopathies originating from mutations in different genes to be grouped together due to similar physical manifestations, and conversely myopathies resulting from mutations in the same gene to be considered separately due to disparate symptoms. Herein, we focus on an early onset myopathy linked to inherited or de novo mutations in sarcomeric genes that is characterized by muscle weakness, hypotonia and tremor, and further highlight that it may constitute a new form of myopathy, with tremor as its defining feature. Based on recent reports, we also discuss the possible myogenic origin of the tremor that may start at the level of the sarcomere due to structural and/or contractile alterations occurring as a result of the identified mutations. It is our hope that establishment of this form of myopathy accompanied by myogenic tremor as a new disease entity will have important diagnostic and therapeutic implications.
Collapse
|
23
|
Wang Z, Feng X, Li SC. SCDevDB: A Database for Insights Into Single-Cell Gene Expression Profiles During Human Developmental Processes. Front Genet 2019; 10:903. [PMID: 31611909 PMCID: PMC6775478 DOI: 10.3389/fgene.2019.00903] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 08/26/2019] [Indexed: 11/19/2022] Open
Abstract
Single-cell RNA-seq studies profile thousands of cells in developmental processes. Current databases for human single-cell expression atlas only provide search and visualize functions for a selected gene in specific cell types or subpopulations. These databases are limited to technical properties or visualization of single-cell RNA-seq data without considering the biological relations of their collected cell groups. Here, we developed a database to investigate single-cell gene expression profiling during different developmental pathways (SCDevDB). In this database, we collected 10 human single-cell RNA-seq datasets, split these datasets into 176 developmental cell groups, and constructed 24 different developmental pathways. SCDevDB allows users to search the expression profiles of the interested genes across different developmental pathways. It also provides lists of differentially expressed genes during each developmental pathway, T-distributed stochastic neighbor embedding maps showing the relationships between developmental stages based on these differentially expressed genes, Gene Ontology, and Kyoto Encyclopedia of Genes and Genomes analysis results of these differentially expressed genes. This database is freely available at https://scdevdb.deepomics.org
Collapse
Affiliation(s)
- Zishuai Wang
- Department of Computer Science, City University of Hong Kong, Kowloon, Hong Kong
| | - Xikang Feng
- Department of Computer Science, City University of Hong Kong, Kowloon, Hong Kong
| | - Shuai Cheng Li
- Department of Computer Science, City University of Hong Kong, Kowloon, Hong Kong
| |
Collapse
|
24
|
Marttila M, Win W, Al-Ghamdi F, Abdel-Hamid HZ, Lacomis D, Beggs AH. MYL2-associated congenital fiber-type disproportion and cardiomyopathy with variants in additional neuromuscular disease genes; the dilemma of panel testing. Cold Spring Harb Mol Case Stud 2019; 5:mcs.a004184. [PMID: 31127036 PMCID: PMC6672024 DOI: 10.1101/mcs.a004184] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 05/16/2019] [Indexed: 12/13/2022] Open
Abstract
Next-generation sequencing has led to transformative advances in our ability to diagnose rare diseases by simultaneously sequencing dozens, hundreds, or even entire genomes worth of genes to efficiently identify pathogenic mutations. These studies amount to multiple hypothesis testing on a massive scale and not infrequently lead to discovery of multiple genetic variants whose relative contributions to a patient's disease are unclear. Panel testing, in particular, can be problematic because each of the many genes being sequenced might represent a plausible explanation for a given case. We performed targeted gene panel analysis of 43 established neuromuscular disease genes in a patient with congenital fiber-type disproportion (CFTD) and fatal infantile cardiomyopathy. Initial review of variants identified changes in four genes that could be considered relevant candidates to cause this child's disease. Further analysis revealed that two of these are likely benign, but a homozygous frameshift variant in the myosin light chain 2 gene, MYL2, and a heterozygous nonsense mutation in the nebulin gene, NEB, met criteria to be classified as likely pathogenic or pathogenic. Recessive MYL2 mutations are a rare cause of CFTD associated with both skeletal and cardiomyopathy, whereas recessive NEB mutations cause nemaline myopathy. Although the proband's phenotype is likely largely explained by the MYL2 variant, the heterozygous pathogenic NEB variant cannot be ruled out as a contributing factor. This case illustrates the complexity when analyzing large numbers of variants from targeted gene panels in which each of the genes might plausibly contribute to the patient's clinical presentation.
Collapse
Affiliation(s)
- Minttu Marttila
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Wathone Win
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Fouad Al-Ghamdi
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA.,King Fahad Specialist Hospital, Dammam 32253, Saudi Arabia
| | - Hoda Z Abdel-Hamid
- Department of Pediatrics, Child Neurology Division, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, USA.,Pediatric MDA Clinic, Division of Child Neurology, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224, USA
| | - David Lacomis
- Neuromuscular Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA
| | - Alan H Beggs
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
25
|
Stavusis J, Lace B, Schäfer J, Geist J, Inashkina I, Kidere D, Pajusalu S, Wright NT, Saak A, Weinhold M, Haubenberger D, Jackson S, Kontrogianni-Konstantopoulos A, Bönnemann CG. Novel mutations in MYBPC1 are associated with myogenic tremor and mild myopathy. Ann Neurol 2019; 86:129-142. [PMID: 31025394 DOI: 10.1002/ana.25494] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 04/23/2019] [Accepted: 04/25/2019] [Indexed: 01/11/2023]
Abstract
OBJECTIVE To define a distinct, dominantly inherited, mild skeletal myopathy associated with prominent and consistent tremor in two unrelated, three-generation families. METHODS Clinical evaluations as well as exome and panel sequencing analyses were performed in affected and nonaffected members of two families to identify genetic variants segregating with the phenotype. Histological assessment of a muscle biopsy specimen was performed in 1 patient, and quantitative tremor analysis was carried out in 2 patients. Molecular modeling studies and biochemical assays were performed for both mutations. RESULTS Two novel missense mutations in MYBPC1 (p.E248K in family 1 and p.Y247H in family 2) were identified and shown to segregate perfectly with the myopathy/tremor phenotype in the respective families. MYBPC1 encodes slow myosin binding protein-C (sMyBP-C), a modular sarcomeric protein playing structural and regulatory roles through its dynamic interaction with actin and myosin filaments. The Y247H and E248K mutations are located in the NH2 -terminal M-motif of sMyBP-C. Both mutations result in markedly increased binding of the NH2 terminus to myosin, possibly interfering with normal cross-bridge cycling as the first muscle-based step in tremor genesis. The clinical tremor features observed in all mutation carriers, together with the tremor physiology studies performed in family 2, suggest amplification by an additional central loop modulating the clinical tremor phenomenology. INTERPRETATION Here, we link two novel missense mutations in MYBPC1 with a dominant, mild skeletal myopathy invariably associated with a distinctive tremor. The molecular, genetic, and clinical studies are consistent with a unique sarcomeric origin of the tremor, which we classify as "myogenic tremor." ANN NEUROL 2019.
Collapse
Affiliation(s)
- Janis Stavusis
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Baiba Lace
- Latvian Biomedical Research and Study Centre, Riga, Latvia.,Centre Hospitalier Universitaire de Québec, Ville de Québec, QC, Canada
| | - Jochen Schäfer
- Department of Neurology-Uniklinikum CG Carus, Dresden, Germany
| | - Janelle Geist
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD
| | - Inna Inashkina
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Dita Kidere
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Sander Pajusalu
- Department of Clinical Genetics, United Laboratories, Tartu University Hospital, Tartu, Estonia.,Department of Clinical Genetics, Institute of Clinical Medicine, Tartu University, Tartu, Estonia
| | - Nathan T Wright
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, VA
| | - Annika Saak
- Department of Neurology-Uniklinikum CG Carus, Dresden, Germany
| | - Manja Weinhold
- Department of Neurology-Uniklinikum CG Carus, Dresden, Germany
| | - Dietrich Haubenberger
- Clinical Trials Unit, Office of the Clinical Director, NINDS Intramural Research Program, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD
| | - Sandra Jackson
- Department of Neurology-Uniklinikum CG Carus, Dresden, Germany
| | | | - Carsten G Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD
| |
Collapse
|
26
|
Ravenscroft G, Zaharieva IT, Bortolotti CA, Lambrughi M, Pignataro M, Borsari M, Sewry CA, Phadke R, Haliloglu G, Ong R, Goullée H, Whyte T, Consortium UK, Manzur A, Talim B, Kaya U, Osborn DPS, Forrest ARR, Laing NG, Muntoni F. Bi-allelic mutations in MYL1 cause a severe congenital myopathy. Hum Mol Genet 2019; 27:4263-4272. [PMID: 30215711 DOI: 10.1093/hmg/ddy320] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 09/07/2018] [Indexed: 01/26/2023] Open
Abstract
Congenital myopathies are typically characterised by early onset hypotonia, weakness and hallmark features on biopsy. Despite the rapid pace of gene discovery, ∼50% of patients with a congenital myopathy remain without a genetic diagnosis following screening of known disease genes. We performed exome sequencing on two consanguineous probands diagnosed with a congenital myopathy and muscle biopsy showing selective atrophy/hypotrophy or absence of type II myofibres. We identified variants in the gene (MYL1) encoding the skeletal muscle fast-twitch specific myosin essential light chain (ELC) in both probands. A homozygous essential splice acceptor variant (c.479-2A > G, predicted to result in skipping of exon 5 was identified in Proband 1, and a homozygous missense substitution (c.488T>G, p.(Met163Arg)) was identified in Proband 2. Protein modelling of the p.(Met163Arg) substitution predicted it might impede intermolecular interactions that facilitate binding to the IQ domain of myosin heavy chain, thus likely impacting on the structure and functioning of the myosin motor. MYL1 was markedly reduced in skeletal muscle from both probands, suggesting that the missense substitution likely results in an unstable protein. Knock down of myl1 in zebrafish resulted in abnormal morphology, disrupted muscle structure and impaired touch-evoked escape responses, thus confirming that skeletal muscle fast-twitch specific myosin ELC is critical for myofibre development and function. Our data implicate MYL1 as a crucial protein for adequate skeletal muscle function and that MYL1 deficiency is associated with severe congenital myopathy.
Collapse
Affiliation(s)
- Gianina Ravenscroft
- Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia
| | - Irina T Zaharieva
- The Dubowitz Neuromuscular Centre, University College London Great Ormond Street Institute of Child Health & Great Ormond Street Hospital, London, UK
| | - Carlo A Bortolotti
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Matteo Lambrughi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Marcello Pignataro
- Department of Chemical and Geological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Marco Borsari
- Department of Chemical and Geological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Caroline A Sewry
- The Dubowitz Neuromuscular Centre, University College London Great Ormond Street Institute of Child Health & Great Ormond Street Hospital, London, UK
| | - Rahul Phadke
- The Dubowitz Neuromuscular Centre, University College London Great Ormond Street Institute of Child Health & Great Ormond Street Hospital, London, UK
| | - Goknur Haliloglu
- Department of Chemical and Geological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Royston Ong
- Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia
| | - Hayley Goullée
- Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia
| | - Tamieka Whyte
- The Dubowitz Neuromuscular Centre, University College London Great Ormond Street Institute of Child Health & Great Ormond Street Hospital, London, UK
| | | | - Adnan Manzur
- The Dubowitz Neuromuscular Centre, University College London Great Ormond Street Institute of Child Health & Great Ormond Street Hospital, London, UK
| | - Beril Talim
- Pediatric Pathology Unit, Hacettepe University Children's Hospital, Ankara, Turkey
| | - Ulkuhan Kaya
- Department of Pediatric Neurology, Dr. Sami Ulus Maternity and Children's Research and Training Hospital, Ministry of Health, Ankara, Turkey
| | - Daniel P S Osborn
- Cardiovascular and Cell Sciences Institute, St George's University of London, Cranmer Terrace, London SW17 0RE, UK
| | - Alistair R R Forrest
- Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia
| | - Nigel G Laing
- Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia
| | - Francesco Muntoni
- The Dubowitz Neuromuscular Centre, University College London Great Ormond Street Institute of Child Health & Great Ormond Street Hospital, London, UK.,NIHR Great Ormond Street Hospital Biomedical Research Centre, 30 Guilford Street, London WC1N 1EH, UK
| |
Collapse
|
27
|
Kazmierczak K, Liang J, Yuan CC, Yadav S, Sitbon YH, Walz K, Ma W, Irving TC, Cheah JX, Gomes AV, Szczesna-Cordary D. Slow-twitch skeletal muscle defects accompany cardiac dysfunction in transgenic mice with a mutation in the myosin regulatory light chain. FASEB J 2019; 33:3152-3166. [PMID: 30365366 PMCID: PMC6404564 DOI: 10.1096/fj.201801402r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 10/01/2018] [Indexed: 01/06/2023]
Abstract
Myosin light chain 2 ( MYL2) gene encodes the myosin regulatory light chain (RLC) simultaneously in heart ventricles and in slow-twitch skeletal muscle. Using transgenic mice with cardiac-specific expression of the human R58Q-RLC mutant, we sought to determine whether the hypertrophic cardiomyopathy phenotype observed in papillary muscles (PMs) of R58Q mice is also manifested in slow-twitch soleus (SOL) muscles. Skinned SOL muscles and ventricular PMs of R58Q animals exhibited lower contractile force that was not observed in the fast-twitch extensor digitorum longus muscles of R58Q vs. wild-type-RLC mice, but mutant animals did not display gross muscle weakness in vivo. Consistent with SOL muscle abnormalities in R58Q vs. wild-type mice, myosin ATPase staining revealed a decreased proportion of fiber type I/type II only in SOL muscles but not in the extensor digitorum longus muscles. The similarities between SOL muscles and PMs of R58Q mice were further supported by quantitative proteomics. Differential regulation of proteins involved in energy metabolism, cell-cell interactions, and protein-protein signaling was concurrently observed in the hearts and SOL muscles of R58Q mice. In summary, even though R58Q expression was restricted to the heart of mice, functional similarities were clearly observed between the hearts and slow-twitch skeletal muscle, suggesting that MYL2 mutated models of hypertrophic cardiomyopathy may be useful research tools to study the molecular, structural, and energetic mechanisms of cardioskeletal myopathy associated with myosin RLC.-Kazmierczak, K., Liang, J., Yuan, C.-C., Yadav, S., Sitbon, Y. H., Walz, K., Ma, W., Irving, T. C., Cheah, J. X., Gomes, A. V., Szczesna-Cordary, D. Slow-twitch skeletal muscle defects accompany cardiac dysfunction in transgenic mice with a mutation in the myosin regulatory light chain.
Collapse
Affiliation(s)
- Katarzyna Kazmierczak
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Jingsheng Liang
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Chen-Ching Yuan
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Sunil Yadav
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Yoel H. Sitbon
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Katherina Walz
- Dr. John T. Macdonald Foundation Department of Human Genetics, John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Weikang Ma
- Department of Biological Sciences, Illinois Institute of Technology, Chicago, Illinois, USA
| | - Thomas C. Irving
- Department of Biological Sciences, Illinois Institute of Technology, Chicago, Illinois, USA
| | - Jenice X. Cheah
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, Davis, California, USA
| | - Aldrin V. Gomes
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, Davis, California, USA
| | - Danuta Szczesna-Cordary
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
28
|
Yadav S, Sitbon YH, Kazmierczak K, Szczesna-Cordary D. Hereditary heart disease: pathophysiology, clinical presentation, and animal models of HCM, RCM, and DCM associated with mutations in cardiac myosin light chains. Pflugers Arch 2019; 471:683-699. [PMID: 30706179 DOI: 10.1007/s00424-019-02257-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 12/26/2018] [Accepted: 01/13/2019] [Indexed: 02/07/2023]
Abstract
Genetic cardiomyopathies, a group of cardiovascular disorders based on ventricular morphology and function, are among the leading causes of morbidity and mortality worldwide. Such genetically driven forms of hypertrophic (HCM), dilated (DCM), and restrictive (RCM) cardiomyopathies are chronic, debilitating diseases that result from biomechanical defects in cardiac muscle contraction and frequently progress to heart failure (HF). Locus and allelic heterogeneity, as well as clinical variability combined with genetic and phenotypic overlap between different cardiomyopathies, have challenged proper clinical prognosis and provided an incentive for identification of pathogenic variants. This review attempts to provide an overview of inherited cardiomyopathies with a focus on their genetic etiology in myosin regulatory (RLC) and essential (ELC) light chains, which are EF-hand protein family members with important structural and regulatory roles. From the clinical discovery of cardiomyopathy-linked light chain mutations in patients to an array of exploratory studies in animals, and reconstituted and recombinant systems, we have summarized the current state of knowledge on light chain mutations and how they induce physiological disease states via biochemical and biomechanical alterations at the molecular, tissue, and organ levels. Cardiac myosin RLC phosphorylation and the N-terminus ELC have been discussed as two important emerging modalities with important implications in the regulation of myosin motor function, and thus cardiac performance. A comprehensive understanding of such triggers is absolutely necessary for the development of target-specific rescue strategies to ameliorate or reverse the effects of myosin light chain-related inherited cardiomyopathies.
Collapse
MESH Headings
- Animals
- Cardiomyopathy, Dilated/etiology
- Cardiomyopathy, Dilated/genetics
- Cardiomyopathy, Dilated/pathology
- Cardiomyopathy, Hypertrophic/etiology
- Cardiomyopathy, Hypertrophic/genetics
- Cardiomyopathy, Hypertrophic/pathology
- Cardiomyopathy, Restrictive/etiology
- Cardiomyopathy, Restrictive/genetics
- Cardiomyopathy, Restrictive/pathology
- Disease Models, Animal
- Humans
- Mutation
- Myosin Light Chains/genetics
Collapse
Affiliation(s)
- Sunil Yadav
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, 1600 NW 10th Ave., Miami, FL, 33136, USA
| | - Yoel H Sitbon
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, 1600 NW 10th Ave., Miami, FL, 33136, USA
| | - Katarzyna Kazmierczak
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, 1600 NW 10th Ave., Miami, FL, 33136, USA
| | - Danuta Szczesna-Cordary
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, 1600 NW 10th Ave., Miami, FL, 33136, USA.
| |
Collapse
|
29
|
Translating emerging molecular genetic insights into clinical practice in inherited cardiomyopathies. J Mol Med (Berl) 2018; 96:993-1024. [PMID: 30128729 DOI: 10.1007/s00109-018-1685-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 07/22/2018] [Accepted: 08/08/2018] [Indexed: 12/19/2022]
Abstract
Cardiomyopathies are primarily genetic disorders of the myocardium associated with higher risk of life-threatening cardiac arrhythmias, heart failure, and sudden cardiac death. The evolving knowledge in genomic medicine during the last decade has reshaped our understanding of cardiomyopathies as diseases of multifactorial nature and complex pathophysiology. Genetic testing in cardiomyopathies has subsequently grown from primarily a research tool into an essential clinical evaluation piece with important clinical implications for patients and their families. The purpose of this review is to provide with a contemporary insight into the implications of genetic testing in diagnosis, therapy, and prognosis of patients with inherited cardiomyopathies. Here, we summarize the contemporary knowledge on genotype-phenotype correlations in inherited cardiomyopathies and highlight the recent significant achievements in the field of translational cardiovascular genetics.
Collapse
|
30
|
Wang L, Geist J, Grogan A, Hu LYR, Kontrogianni-Konstantopoulos A. Thick Filament Protein Network, Functions, and Disease Association. Compr Physiol 2018; 8:631-709. [PMID: 29687901 PMCID: PMC6404781 DOI: 10.1002/cphy.c170023] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Sarcomeres consist of highly ordered arrays of thick myosin and thin actin filaments along with accessory proteins. Thick filaments occupy the center of sarcomeres where they partially overlap with thin filaments. The sliding of thick filaments past thin filaments is a highly regulated process that occurs in an ATP-dependent manner driving muscle contraction. In addition to myosin that makes up the backbone of the thick filament, four other proteins which are intimately bound to the thick filament, myosin binding protein-C, titin, myomesin, and obscurin play important structural and regulatory roles. Consistent with this, mutations in the respective genes have been associated with idiopathic and congenital forms of skeletal and cardiac myopathies. In this review, we aim to summarize our current knowledge on the molecular structure, subcellular localization, interacting partners, function, modulation via posttranslational modifications, and disease involvement of these five major proteins that comprise the thick filament of striated muscle cells. © 2018 American Physiological Society. Compr Physiol 8:631-709, 2018.
Collapse
Affiliation(s)
- Li Wang
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | - Janelle Geist
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | - Alyssa Grogan
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | - Li-Yen R. Hu
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | | |
Collapse
|
31
|
Veltri T, Landim-Vieira M, Parvatiyar MS, Gonzalez-Martinez D, Dieseldorff Jones KM, Michell CA, Dweck D, Landstrom AP, Chase PB, Pinto JR. Hypertrophic Cardiomyopathy Cardiac Troponin C Mutations Differentially Affect Slow Skeletal and Cardiac Muscle Regulation. Front Physiol 2017; 8:221. [PMID: 28473771 PMCID: PMC5397416 DOI: 10.3389/fphys.2017.00221] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 03/27/2017] [Indexed: 12/22/2022] Open
Abstract
Mutations in TNNC1-the gene encoding cardiac troponin C (cTnC)-that have been associated with hypertrophic cardiomyopathy (HCM) and cardiac dysfunction may also affect Ca2+-regulation and function of slow skeletal muscle since the same gene is expressed in both cardiac and slow skeletal muscle. Therefore, we reconstituted rabbit soleus fibers and bovine masseter myofibrils with mutant cTnCs (A8V, C84Y, E134D, and D145E) associated with HCM to investigate their effects on contractile force and ATPase rates, respectively. Previously, we showed that these HCM cTnC mutants, except for E134D, increased the Ca2+ sensitivity of force development in cardiac preparations. In the current study, an increase in Ca2+ sensitivity of isometric force was only observed for the C84Y mutant when reconstituted in soleus fibers. Incorporation of cTnC C84Y in bovine masseter myofibrils reduced the ATPase activity at saturating [Ca2+], whereas, incorporation of cTnC D145E increased the ATPase activity at inhibiting and saturating [Ca2+]. We also tested whether reconstitution of cardiac fibers with troponin complexes containing the cTnC mutants and slow skeletal troponin I (ssTnI) could emulate the slow skeletal functional phenotype. Reconstitution of cardiac fibers with troponin complexes containing ssTnI attenuated the Ca2+ sensitization of isometric force when cTnC A8V and D145E were present; however, it was enhanced for C84Y. In summary, although the A8V and D145E mutants are present in both muscle types, their functional phenotype is more prominent in cardiac muscle than in slow skeletal muscle, which has implications for the protein-protein interactions within the troponin complex. The C84Y mutant warrants further investigation since it drastically alters the properties of both muscle types and may account for the earlier clinical onset in the proband.
Collapse
Affiliation(s)
- Tiago Veltri
- Department of Biomedical Sciences, Florida State University College of MedicineTallahassee, FL, USA
| | - Maicon Landim-Vieira
- Department of Biomedical Sciences, Florida State University College of MedicineTallahassee, FL, USA
| | - Michelle S. Parvatiyar
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of MedicineMiami, FL, USA
| | - David Gonzalez-Martinez
- Department of Biomedical Sciences, Florida State University College of MedicineTallahassee, FL, USA
| | | | - Clara A. Michell
- Department of Biomedical Sciences, Florida State University College of MedicineTallahassee, FL, USA
| | - David Dweck
- Department of Biomedical Sciences, Florida State University College of MedicineTallahassee, FL, USA
| | - Andrew P. Landstrom
- Section of Pediatric Cardiology, Department of Pediatrics, Baylor College of MedicineHouston, TX, USA
| | - P. Bryant Chase
- Department of Biological Science, Florida State UniversityTallahassee, FL, USA
| | - Jose R. Pinto
- Department of Biomedical Sciences, Florida State University College of MedicineTallahassee, FL, USA
| |
Collapse
|
32
|
Zhou Z, Huang W, Liang J, Szczesna-Cordary D. Molecular and Functional Effects of a Splice Site Mutation in the MYL2 Gene Associated with Cardioskeletal Myopathy and Early Cardiac Death in Infants. Front Physiol 2016; 7:240. [PMID: 27378946 PMCID: PMC4911367 DOI: 10.3389/fphys.2016.00240] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 06/03/2016] [Indexed: 12/26/2022] Open
Abstract
The homozygous appearance of the intronic mutation (IVS6-1) in the MYL2 gene encoding for myosin ventricular/slow-twitch skeletal regulatory light chain (RLC) was recently linked to the development of slow skeletal muscle fiber type I hypotrophy and early cardiac death. The IVS6-1 (c403-1G>C) mutation resulted from a cryptic splice site in MYL2 causing a frameshift and replacement of the last 32 codons by 19 different amino acids in the RLC mutant protein. Infants who were IVS6-1+∕+-positive died between 4 and 6 months of age due to cardiomyopathy and heart failure. In this report we have investigated the molecular mechanism and functional consequences associated with the IVS6-1 mutation using recombinant human cardiac IVS6-1 and wild-type (WT) RLC proteins. Recombinant proteins were reconstituted into RLC-depleted porcine cardiac muscle preparations and subjected to enzymatic and functional assays. IVS6-1-RLC showed decreased binding to the myosin heavy chain (MHC) compared with WT, and IVS6-1-reconstituted myosin displayed reduced binding to actin in rigor. The IVS6-1 myosin demonstrated a significantly lower Vmax of the actin-activated myosin ATPase activity compared with WT. In stopped-flow experiments, IVS6-1 myosin showed slower kinetics of the ATP induced dissociation of the acto-myosin complex and a significantly reduced slope of the kobs-[MgATP] relationship compared to WT. In skinned porcine cardiac muscles, RLC-depleted and IVS6-1 reconstituted muscle strips displayed a significant decrease in maximal contractile force and a significantly increased Ca2+ sensitivity, both hallmarks of hypertrophic cardiomyopathy-associated mutations in MYL2. Our results showed that the amino-acid changes in IVS6-1 were sufficient to impose significant conformational alterations in the RLC protein and trigger a series of abnormal protein-protein interactions in the cardiac muscle sarcomere. Notably, the mutation disrupted the RLC-MHC interaction and the steady-state and kinetics of the acto-myosin interaction. Specifically, slower myosin cross-bridge turnover rates and slower second-order MgATP binding rates of acto-myosin interactions were observed in IVS6-1 vs. WT reconstituted cardiac preparations. Our in vitro results suggest that when placed in vivo, IVS6-1 may lead to cardiomyopathy and early death of homozygous infants by severely compromising the ability of myosin to develop contractile force and maintain normal systolic and diastolic cardiac function.
Collapse
Affiliation(s)
- Zhiqun Zhou
- Department of Molecular and Cellular Pharmacology, University of Miami Leonard M. Miller School of Medicine Miami, FL, USA
| | - Wenrui Huang
- Department of Molecular and Cellular Pharmacology, University of Miami Leonard M. Miller School of Medicine Miami, FL, USA
| | - Jingsheng Liang
- Department of Molecular and Cellular Pharmacology, University of Miami Leonard M. Miller School of Medicine Miami, FL, USA
| | - Danuta Szczesna-Cordary
- Department of Molecular and Cellular Pharmacology, University of Miami Leonard M. Miller School of Medicine Miami, FL, USA
| |
Collapse
|
33
|
Zhao T, Huang X, Xia Y. Human heart cell proteins interacting with a C-terminally truncated 2A protein of coxsackie B3 virus: identification by the yeast two-hybrid system. Virus Genes 2016; 52:172-8. [PMID: 26781950 DOI: 10.1007/s11262-015-1270-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 12/08/2015] [Indexed: 11/27/2022]
Abstract
Protein 2A is a non-structural protein of coxsackievirus B3 (CVB3), an important human pathogen that can cause a variety of human diseases. Protein 2A not only participates in viral life cycle, but also regulates host cell functions; however, the underlying mechanisms remain poorly understood. In order to better understand the molecular mechanisms of CVB3 2A's function, the yeast two-hybrid (Y2H) system was adopted to screen for CVB3 2A interactive proteins in the human heart cDNA library. Full-length 2A shows strong transcriptional activity in yeast cells, which interferes with the application of Y2H system; therefore, a series of 2A deletion mutants were constructed. Analysis of transcriptional self-activation revealed that 2A lost its transcriptional activity after truncation of 60 amino acids (aa) at the N-terminus or deletion of 17 aa at the C-terminus. Choosing the 2A mutant with 17 aa deletion at the C-terminus as the bait protein, four interactive cellular proteins were identified, including TIMP4, MYL2, COX7C, and ENO1. These proteins are mostly related to protein degradation and metabolism. Although the interactions detected by the Y2H system should be considered as preliminary results, the finding of proteins translated from a human heart cDNA library that interacts with the CVB3 2A will stimulate experiments testing the reactivity of a translational mixture derived from that library with full-length 2A protein, followed by co-immunoprecipitation studies.
Collapse
Affiliation(s)
- Tiansheng Zhao
- School of Pharmacy, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xiaotian Huang
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Yanhua Xia
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
34
|
Cho ER, Jee YH, Kim SW, Sull JW. Effect of obesity on the association between MYL2 (rs3782889) and high-density lipoprotein cholesterol among Korean men. J Hum Genet 2016; 61:405-9. [PMID: 26763873 DOI: 10.1038/jhg.2015.165] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 11/30/2015] [Accepted: 12/09/2015] [Indexed: 01/01/2023]
Abstract
High-density lipoprotein (HDL) cholesterol levels are associated with a decreased risk of coronary artery disease. Several genome-wide association studies that have examined HDL cholesterol levels have implicated myosin light chain 2 regulatory cardiac slow (MYL2) as a possible causal factor. Herein, the association between the rs3782889 single-nucleotide polymorphism (SNP) in the MYL2 gene and HDL cholesterol levels was tested in the Korean population. A total of 4294 individuals were included in a replication study with MYL2 SNP rs3782889. SNP rs3782889 in the MYL2 gene was associated with mean HDL cholesterol level (effect per allele, -1.055 mg dl(-1), P=0.0005). Subjects with the CT/CC genotype had a 1.43-fold (range 1.19-1.73-fold) higher risk of an abnormal HDL cholesterol level (<40 mg dl(-1)) than subjects with the TT genotype. When analyzed by sex, the MYL2 association was stronger in men than that in women. When analyzed by body mass index (BMI), the MYL2 association was much stronger in male subjects with BMI ⩾26.44 kg m(-2) (odds ratio (OR)=2.68; 95% confidence interval (CI)=1.87-3.84; P<0.0001) than that in male subjects with BMI <26.44 kg m(-2). When compared with subjects having the TT genotype and BMI <26.44 kg m(-2), ORs (95% CI) were 3.30 (2.41-4.50) in subjects having the CT/CC genotype and BMI ⩾26.44 kg m(-2) (P for interaction <0.0001). Our results clearly demonstrate that genetic variants in MYL2 influence HDL cholesterol levels in Korean obese male subjects.
Collapse
Affiliation(s)
- Eo Rin Cho
- Department of Epidemiology and Health Promotion, Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea
| | - Yon Ho Jee
- Department of Statistics, Sookmyung Women's University, Seoul, Korea
| | - Sang Won Kim
- Department of Natural Healing, Dongbang Culture Graduate University, Seoul, Korea
| | - Jae Woong Sull
- Department of Biomedical Laboratory Science, College of Health Sciences, Eulji University, Seongnam, Korea
| |
Collapse
|
35
|
Xia J, Zhang Y, Xin L, Kong S, Chen Y, Yang S, Li K. Global Transcriptomic Profiling of Cardiac Hypertrophy and Fatty Heart Induced by Long-Term High-Energy Diet in Bama Miniature Pigs. PLoS One 2015; 10:e0132420. [PMID: 26161779 PMCID: PMC4498776 DOI: 10.1371/journal.pone.0132420] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 06/13/2015] [Indexed: 12/31/2022] Open
Abstract
A long-term high-energy diet affects human health and leads to obesity and metabolic syndrome in addition to cardiac steatosis and hypertrophy. Ectopic fat accumulation in the heart has been demonstrated to be a risk factor for heart disorders, but the molecular mechanism of heart disease remains largely unknown. Bama miniature pigs were fed a high-fat, high-sucrose diet (HFHSD) for 23 months. These pigs developed symptoms of metabolic syndrome and showed cardiac steatosis and hypertrophy with a greatly increased body weight (2.73-fold, P<0.01), insulin level (4.60-fold, P<0.01), heart weight (1.82-fold, P<0.05) and heart volume (1.60-fold, P<0.05) compared with the control pigs. To understand the molecular mechanisms of cardiac steatosis and hypertrophy, nine pig heart cRNA samples were hybridized to porcine GeneChips. Microarray analyses revealed that 1,022 genes were significantly differentially expressed (P<0.05, ≥1.5-fold change), including 591 up-regulated and 431 down-regulated genes in the HFHSD group relative to the control group. KEGG analysis indicated that the observed heart disorder involved the signal transduction-related MAPK, cytokine, and PPAR signaling pathways, energy metabolism-related fatty acid and oxidative phosphorylation signaling pathways, heart function signaling-related focal adhesion, axon guidance, hypertrophic cardiomyopathy and actin cytoskeleton signaling pathways, inflammation and apoptosis pathways, and others. Quantitative RT-PCR assays identified several important differentially expressed heart-related genes, including STAT3, ACSL4, ATF4, FADD, PPP3CA, CD74, SLA-8, VCL, ACTN2 and FGFR1, which may be targets of further research. This study shows that a long-term, high-energy diet induces obesity, cardiac steatosis, and hypertrophy and provides insights into the molecular mechanisms of hypertrophy and fatty heart to facilitate further research.
Collapse
Affiliation(s)
- Jihan Xia
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, P. R. China
| | - Yuanyuan Zhang
- College of Veterinary Medicine, China Agricultural University, Beijing, P. R. China
| | - Leilei Xin
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, P. R. China
| | - Siyuan Kong
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, P. R. China
| | - Yaoxing Chen
- College of Veterinary Medicine, China Agricultural University, Beijing, P. R. China
| | - Shulin Yang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, P. R. China
- * E-mail:
| | - Kui Li
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, P. R. China
- Agricultural Genomes Institute at Shenzhen, CAAS, Shenzhen, 518120, P.R. China
| |
Collapse
|
36
|
Tardiff JC, Carrier L, Bers DM, Poggesi C, Ferrantini C, Coppini R, Maier LS, Ashrafian H, Huke S, van der Velden J. Targets for therapy in sarcomeric cardiomyopathies. Cardiovasc Res 2015; 105:457-70. [PMID: 25634554 DOI: 10.1093/cvr/cvv023] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
To date, no compounds or interventions exist that treat or prevent sarcomeric cardiomyopathies. Established therapies currently improve the outcome, but novel therapies may be able to more fundamentally affect the disease process and course. Investigations of the pathomechanisms are generating molecular insights that can be useful for the design of novel specific drugs suitable for clinical use. As perturbations in the heart are stage-specific, proper timing of drug treatment is essential to prevent initiation and progression of cardiac disease in mutation carrier individuals. In this review, we emphasize potential novel therapies which may prevent, delay, or even reverse hypertrophic cardiomyopathy caused by sarcomeric gene mutations. These include corrections of genetic defects, altered sarcomere function, perturbations in intracellular ion homeostasis, and impaired myocardial energetics.
Collapse
Affiliation(s)
- Jil C Tardiff
- Department of Medicine and Cellular and Molecular Medicine, University of Arizona, 1656 East Mabel Street, MRB 312, Tucson, AZ 85724-5217, USA
| | - Lucie Carrier
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Donald M Bers
- Department of Pharmacology, University of California, Davis, CA, USA
| | - Corrado Poggesi
- Center of Molecular Medicine and Applied Biophysics (CIMMBA), University of Florence, Florence, Italy
| | - Cecilia Ferrantini
- Center of Molecular Medicine and Applied Biophysics (CIMMBA), University of Florence, Florence, Italy
| | - Raffaele Coppini
- Center of Molecular Medicine and Applied Biophysics (CIMMBA), University of Florence, Florence, Italy
| | - Lars S Maier
- Klinik und Poliklinik für Innere Medizin II, Universitätsklinikum, Regensburg, Germany
| | - Houman Ashrafian
- Experimental Therapeutics and Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Sabine Huke
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jolanda van der Velden
- Department of Physiology, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands ICIN-Netherlands Heart Institute, Utrecht, the Netherlands
| |
Collapse
|
37
|
Park HJ, Choi YC, Kim SM, Kim SH, Hong YB, Yoon BR, Chung KW, Choi BO. Molecular Genetic Diagnosis of a Bethlem Myopathy Family with an Autosomal-Dominant COL6A1 Mutation, as Evidenced by Exome Sequencing. J Clin Neurol 2014; 11:183-7. [PMID: 25749816 PMCID: PMC4387485 DOI: 10.3988/jcn.2015.11.2.183] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 09/03/2013] [Accepted: 09/06/2013] [Indexed: 01/09/2023] Open
Abstract
Background We describe herein the application of whole exome sequencing (WES) for the molecular genetic diagnosis of a large Korean family with dominantly inherited myopathy. Case Report The affected individuals presented with slowly progressive proximal weakness and ankle contracture. They were initially diagnosed with limb-girdle muscular dystrophy (LGMD) based on clinical and pathologic features. However, WES and subsequent capillary sequencing identified a pathogenic splicing-site mutation (c.1056+1G>A) in COL6A1, which was previously reported to be an underlying cause of Bethlem myopathy. After identification of the genetic cause of the disease, careful neurologic examination revealed subtle contracture of the interphalangeal joint in the affected members, which is a characteristic sign of Bethlem myopathy. Therefore, we revised the original diagnosis from LGMD to Bethlem myopathy. Conclusions This is the first report of identification of COL6A1-mediated Bethlem myopathy in Korea, and indicates the utility of WES for the diagnosis of muscular dystrophy.
Collapse
Affiliation(s)
- Hyung Jun Park
- Department of Neurology, Mokdong Hospital, Ewha Womans University School of Medicine, Seoul, Korea
| | - Young Chul Choi
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Seung Min Kim
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Se Hoon Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
| | - Young Bin Hong
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Bo Ram Yoon
- Department of Biological Science, Kongju National University, Gongju, Korea
| | - Ki Wha Chung
- Department of Biological Science, Kongju National University, Gongju, Korea.
| | - Byung Ok Choi
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
| |
Collapse
|
38
|
Genome-wide DNA methylation profiles and their relationships with mRNA and the microRNA transcriptome in bovine muscle tissue (Bos taurine). Sci Rep 2014; 4:6546. [PMID: 25306978 PMCID: PMC4194443 DOI: 10.1038/srep06546] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 09/15/2014] [Indexed: 12/21/2022] Open
Abstract
DNA methylation is a key epigenetic modification in mammals and plays important roles in muscle development. We sampled longissimus dorsi muscle (LDM) from a well-known elite native breed of Chinese Qinchuan cattle living within the same environment but displaying distinct skeletal muscle at the fetal and adult stages. We generated and provided a genome-wide landscape of DNA methylomes and their relationship with mRNA and miRNA for fetal and adult muscle studies. Integration analysis revealed a total of 77 and 1,054 negatively correlated genes with methylation in the promoter and gene body regions, respectively, in both the fetal and adult bovine libraries. Furthermore, we identified expression patterns of high-read genes that exhibit a negative correlation between methylation and expression from nine different tissues at multiple developmental stages of bovine muscle-related tissue or organs. In addition, we validated the MeDIP-Seq results by bisulfite sequencing PCR (BSP) in some of the differentially methylated promoters. Together, these results provide valuable data for future biomedical research and genomic and epigenomic studies of bovine skeletal muscle that may help uncover the molecular basis underlying economically valuable traits in cattle. This comprehensive map also provides a solid basis for exploring the epigenetic mechanisms of muscle growth and development.
Collapse
|
39
|
Li H, Wang X, Mao Y, Hu R, Xu W, Lei Z, Zhou N, Jin L, Guo T, Li Z, Irwin DM, Niu G, Tan H. Long term liver specific glucokinase gene defect induced diabetic cardiomyopathy by up regulating NADPH oxidase and down regulating insulin receptor and p-AMPK. Cardiovasc Diabetol 2014; 13:24. [PMID: 24447392 PMCID: PMC3901845 DOI: 10.1186/1475-2840-13-24] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 01/17/2014] [Indexed: 01/12/2023] Open
Abstract
Background The liver-specific glucokinase knockout (gckw/–) mouse experiences long-term hyperglycemia and insulin resistance. This study was designed to evaluate the functional and structural changes in the myocardium of 60 week-old gckw/– mice, and to investigate the effect of rosiglitazone on the myocardium in this model. Methods 60 week-old gckw/– mice were randomly divided into 3 groups: gckw/–, gckw/– mice treated with insulin (1 U/kg) and gckw/– mice treated with rosiglitazone (18 mg/kg). Insulin or rosiglitazone treatment was for 4 weeks. Gckw/w litermates were used as controls. Echocardiography, electrocardiogram, biochemical, histopathological, ultrastructural, real time PCR and Western blot studies were performed to examine for structural and functional changes. Results Long-term liver-specific gck knockout in mice elicits hyperglycaemia and insulin resistance. Compared to age matched gckw/w mice, 60 week-old gckw/– mice showed decreased LV internal dimension, increased posterior wall thickness, lengthened PR and QRS intervals, up-regulated MLC2 protein expression, decreased SOD activity, increased MDA levels and up-regulated Cyba mRNA. Morphological studies revealed that there was an increase in the amount of PAS and Masson positively stained material, as did the number and proportion of the cell occupied by mitochondria in the gckw/– mice. Western blot analysis revealed that the levels of the insulin receptor, Akt, phosphorylated AMPK beta and phosphorylated ACC were reduced in gckw/– mice. These effects were partly attenuated or ablated by treatment with rosiglitazone. Conclusions Our results indicate that changes in the myocardium occur in the liver-specific glucokinase knockout mouse and suggest that reduced glucokinase expression in the liver may induce diabetic cardiomyopathy by up regulating NADPH oxidase and down regulating insulin receptor and p-AMPK protein levels. Rosiglitazone treatment may protect against diabetic cardiomyopathy by altering the levels of a set of proteins involved in cardiac damage.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Gang Niu
- Department of Pharmacology, Health Science Center, Peking University, 38 Xue Yuan Road, Beijing 100191, China.
| | | |
Collapse
|
40
|
Zhang F, Liu C, Xu Y, Qi G, Yuan G, Cheng Z, Wang J, Wang G, Wang Z, Zhu W, Zhou Z, Zhao X, Tian L, Jin C, Yuan J, Zhang G, Chen Y, Wang L, Lu T, Yan H, Ruan Y, Yue W, Zhang D. A two-stage association study suggests BRAP as a susceptibility gene for schizophrenia. PLoS One 2014; 9:e86037. [PMID: 24454952 PMCID: PMC3893271 DOI: 10.1371/journal.pone.0086037] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 12/04/2013] [Indexed: 01/05/2023] Open
Abstract
Schizophrenia (SZ) is a neurodevelopmental disorder in which altered immune function typically plays an important role in mediating the effect of environmental insults and regulation of inflammation. The breast cancer suppressor protein associated protein (BRAP) is suggested to exert vital effects in neurodevelopment by modulating the mitogen-activated protein kinase cascade and inflammation signaling. To explore the possible role of BRAP in SZ, we conducted a two-stage study to examine the association of BRAP polymorphisms with SZ in the Han Chinese population. In stage one, we screened SNPs in BRAP from our GWAS data, which detected three associated SNPs, with rs3782886 being the most significant one (P = 2.31E-6, OR = 0.67). In stage two, we validated these three SNPs in an independently collected population including 1957 patients and 1509 controls, supporting the association of rs3782886 with SZ (P = 1.43E-6, OR = 0.73). Furthermore, cis-eQTL analysis indicates that rs3782886 genotypes are associated with mRNA levels of aldehyde dehydrogenase 2 family (ALDH2) (P = 0.0039) and myosin regulatory light chain 2 (MYL2) (P < 1.0E-4). Our data suggest that the BRAP gene may confer vulnerability for SZ in Han Chinese population, adding further evidence for the involvement of developmental and/or neuroinflammatory cascades in the illness.
Collapse
Affiliation(s)
- Fuquan Zhang
- Wuxi Mental Health Center of Nanjing Medical University, Wuxi, Jiangsu Province, China
- Key Laboratory of Mental Health, Ministry of Health, Institute of Mental Health, The Sixth Hospital, Peking University, Beijing, China
| | - Chenxing Liu
- Key Laboratory of Mental Health, Ministry of Health, Institute of Mental Health, The Sixth Hospital, Peking University, Beijing, China
| | - Yong Xu
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Guoyang Qi
- Wuxi Mental Health Center of Nanjing Medical University, Wuxi, Jiangsu Province, China
| | - Guozhen Yuan
- Wuxi Mental Health Center of Nanjing Medical University, Wuxi, Jiangsu Province, China
| | - Zaohuo Cheng
- Wuxi Mental Health Center of Nanjing Medical University, Wuxi, Jiangsu Province, China
| | - Jidong Wang
- Wuxi Mental Health Center of Nanjing Medical University, Wuxi, Jiangsu Province, China
| | - Guoqiang Wang
- Wuxi Mental Health Center of Nanjing Medical University, Wuxi, Jiangsu Province, China
| | - Zhiqiang Wang
- Wuxi Mental Health Center of Nanjing Medical University, Wuxi, Jiangsu Province, China
| | - Wei Zhu
- Wuxi Mental Health Center of Nanjing Medical University, Wuxi, Jiangsu Province, China
| | - Zhenhe Zhou
- Wuxi Mental Health Center of Nanjing Medical University, Wuxi, Jiangsu Province, China
| | - Xingfu Zhao
- Wuxi Mental Health Center of Nanjing Medical University, Wuxi, Jiangsu Province, China
| | - Lin Tian
- Wuxi Mental Health Center of Nanjing Medical University, Wuxi, Jiangsu Province, China
| | - Chunhui Jin
- Wuxi Mental Health Center of Nanjing Medical University, Wuxi, Jiangsu Province, China
| | - Janmin Yuan
- Wuxi Mental Health Center of Nanjing Medical University, Wuxi, Jiangsu Province, China
| | - Guofu Zhang
- Wuxi Mental Health Center of Nanjing Medical University, Wuxi, Jiangsu Province, China
| | - Yaguang Chen
- Key Laboratory of Mental Health, Ministry of Health, Institute of Mental Health, The Sixth Hospital, Peking University, Beijing, China
| | - Lifang Wang
- Key Laboratory of Mental Health, Ministry of Health, Institute of Mental Health, The Sixth Hospital, Peking University, Beijing, China
| | - Tianlan Lu
- Key Laboratory of Mental Health, Ministry of Health, Institute of Mental Health, The Sixth Hospital, Peking University, Beijing, China
| | - Hao Yan
- Key Laboratory of Mental Health, Ministry of Health, Institute of Mental Health, The Sixth Hospital, Peking University, Beijing, China
| | - Yanyan Ruan
- Key Laboratory of Mental Health, Ministry of Health, Institute of Mental Health, The Sixth Hospital, Peking University, Beijing, China
| | - Weihua Yue
- Key Laboratory of Mental Health, Ministry of Health, Institute of Mental Health, The Sixth Hospital, Peking University, Beijing, China
- * E-mail: (DZ); (WY)
| | - Dai Zhang
- Key Laboratory of Mental Health, Ministry of Health, Institute of Mental Health, The Sixth Hospital, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
- * E-mail: (DZ); (WY)
| |
Collapse
|