1
|
Zhao J, Xu Y. PITX1 plays essential functions in cancer. Front Oncol 2023; 13:1253238. [PMID: 37841446 PMCID: PMC10570508 DOI: 10.3389/fonc.2023.1253238] [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: 07/05/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023] Open
Abstract
PITX1, also known as the pituitary homeobox 1 gene, has emerged as a key regulator in animal growth and development, attracting significant research attention. Recent investigations have revealed the implication of dysregulated PITX1 expression in tumorigenesis, highlighting its involvement in cancer development. Notably, PITX1 interacts with p53 and exerts control over crucial cellular processes including cell cycle progression, apoptosis, and chemotherapy resistance. Its influence extends to various tumors, such as esophageal, colorectal, gastric, and liver cancer, contributing to tumor progression and metastasis. Despite its significance, a comprehensive review examining PITX1's role in oncology remains lacking. This review aims to address this gap by providing a comprehensive overview of PITX1 in different cancer types, with a particular focus on its clinicopathological significance.
Collapse
Affiliation(s)
- Jingpu Zhao
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, Shandong, China
| | - Yongfeng Xu
- Abdominal Oncology Ward, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
2
|
Sun D, Ding Z, Hai Y, Cheng Y. Advances in epigenetic research of adolescent idiopathic scoliosis and congenital scoliosis. Front Genet 2023; 14:1211376. [PMID: 37564871 PMCID: PMC10411889 DOI: 10.3389/fgene.2023.1211376] [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: 04/24/2023] [Accepted: 06/30/2023] [Indexed: 08/12/2023] Open
Abstract
Scoliosis is a three-dimensional structural deformity of the spine; more than 80% of scoliosis has no specific pathogenesis but is understood to be closely related to genetic, hormonal, and environmental factors. In recent years, the epigenetic alterations observed in scoliosis have been analyzed in numerous studies to determine the pathogenesis and progression of this condition, however, there is currently no comprehensive review of the epigenetic factors to date. We searched PubMed, Embase, and Web of Science databases for relative studies without language and date restrictions in March 2023. Twenty-five studies were included in this review and analyzed from the four main aspects of epigenetic alteration: DNA methylation, non-coding RNAs, histone modifications, and chromatin remodeling. The relationship between DNA methylation, non-coding RNAs, and scoliosis was considerably reported in the literature, and the corresponding related signaling pathways and novel biomarkers observed in scoliosis provide insights into innovative prevention and treatment strategies. However, the role of histone modifications is rarely reported in scoliosis, and few studies have investigated the relationship between scoliosis and chromatin remodeling. Therefore, these related fields need to be further explored to elucidate the overall effects of epigenetics in scoliosis.
Collapse
Affiliation(s)
| | | | - Yong Hai
- Department of Orthopedic Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | | |
Collapse
|
3
|
Murphy K, Zhang A, Bittel AJ, Chen YW. Molecular and Phenotypic Changes in FLExDUX4 Mice. J Pers Med 2023; 13:1040. [PMID: 37511653 PMCID: PMC10381554 DOI: 10.3390/jpm13071040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 06/13/2023] [Accepted: 06/16/2023] [Indexed: 07/30/2023] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is caused by the aberrant expression of the double homeobox 4 (DUX4) gene. The FLExDUX4 mouse model carries an inverted human DUX4 transgene which has leaky DUX4 transgene expression at a very low level. No overt muscle pathology was reported before 16 weeks. The purpose of this study is to track and characterize the FLExDUX4 phenotypes for a longer period, up to one year old. In addition, transcriptomic changes in the muscles of 2-month-old mice were investigated using RNA-seq. The results showed that male FLExDUX4 mice developed more severe phenotypes and at a younger age in comparison to the female mice. These include lower body and muscle weight, and muscle weakness measured by grip strength measurements. Muscle pathological changes were observed at older ages, including fibrosis, decreased size of type IIa and IIx myofibers, and the development of aggregates containing TDP-43 in type IIb myofibers. Muscle transcriptomic data identified early molecular changes in biological pathways regulating circadian rhythm and adipogenesis. The study suggests a slow progressive change in molecular and muscle phenotypes in response to the low level of DUX4 expression in the FLExDUX4 mice.
Collapse
Affiliation(s)
- Kelly Murphy
- Institute for Biomedical Sciences, The George Washington University, Washington, DC 20037, USA
| | - Aiping Zhang
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC 20010, USA
| | - Adam J Bittel
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC 20010, USA
| | - Yi-Wen Chen
- Institute for Biomedical Sciences, The George Washington University, Washington, DC 20037, USA
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC 20010, USA
- Department of Genomics and Precision Medicine, School of Medicine and Health Science, The George Washington University, Washington, DC 20037, USA
| |
Collapse
|
4
|
Shao M, Shi K, Zhao Q, Duan Y, Shen Y, Tian J, He K, Li D, Yu M, Lu Y, Tang Y, Feng C. Transcriptome Analysis Reveals the Differentially Expressed Genes Associated with Growth in Guangxi Partridge Chickens. Genes (Basel) 2022; 13:genes13050798. [PMID: 35627183 PMCID: PMC9140345 DOI: 10.3390/genes13050798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/27/2022] [Accepted: 04/27/2022] [Indexed: 02/04/2023] Open
Abstract
The Guangxi Partridge chicken is a well-known chicken breed in southern China with good meat quality, which has been bred as a meat breed to satisfy the increased demand of consumers. Compared with line D whose body weight is maintained at the average of the unselected group, the growth rate and weight of the selected chicken group (line S) increased significantly after breeding for four generations. Herein, transcriptome analysis was performed to identify pivotal genes and signal pathways of selective breeding that contributed to potential mechanisms of growth and development under artificial selection pressure. The average body weight of line S chickens was 1.724 kg at 90 d of age, which showed a significant increase at 90 d of age than line D chickens (1.509 kg), although only the internal organ ratios of lung and kidney changed after standardizing by body weight. The myofiber area and myofiber density of thigh muscles were affected by selection to a greater extent than that of breast muscle. We identified 51, 210, 31, 388, and 100 differentially expressed genes (DEGs) in the hypothalamus, pituitary, breast muscle, thigh muscle, and liver between the two lines, respectively. Several key genes were identified in the hypothalamus-pituitary-muscle axis, such as FST, THSB, PTPRJ, CD36, PITX1, PITX2, AMPD1, PRKAB1, PRKAB2, and related genes for muscle development, which were attached to the cytokine–cytokine receptor interaction signaling pathway, the PPAR signaling pathway, and lipid metabolism. However, signaling molecular pathways and the cell community showed that elevated activity in the liver of line S fowl was mainly involved in focal adhesion, ECM-receptor interaction, cell adhesion molecules, and signal transduction. Collectively, muscle development, lipid metabolism, and several signaling pathways played crucial roles in the improving growth performance of Guangxi Partridge chickens under artificial selection for growth rate. These results support further study of the adaptation of birds under selective pressure.
Collapse
Affiliation(s)
- Minghui Shao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (M.S.); (K.S.); (Q.Z.); (Y.D.); (Y.S.); (J.T.); (K.H.); (D.L.); (M.Y.)
| | - Kai Shi
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (M.S.); (K.S.); (Q.Z.); (Y.D.); (Y.S.); (J.T.); (K.H.); (D.L.); (M.Y.)
| | - Qian Zhao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (M.S.); (K.S.); (Q.Z.); (Y.D.); (Y.S.); (J.T.); (K.H.); (D.L.); (M.Y.)
| | - Ying Duan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (M.S.); (K.S.); (Q.Z.); (Y.D.); (Y.S.); (J.T.); (K.H.); (D.L.); (M.Y.)
| | - Yangyang Shen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (M.S.); (K.S.); (Q.Z.); (Y.D.); (Y.S.); (J.T.); (K.H.); (D.L.); (M.Y.)
| | - Jinjie Tian
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (M.S.); (K.S.); (Q.Z.); (Y.D.); (Y.S.); (J.T.); (K.H.); (D.L.); (M.Y.)
| | - Kun He
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (M.S.); (K.S.); (Q.Z.); (Y.D.); (Y.S.); (J.T.); (K.H.); (D.L.); (M.Y.)
| | - Dongfeng Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (M.S.); (K.S.); (Q.Z.); (Y.D.); (Y.S.); (J.T.); (K.H.); (D.L.); (M.Y.)
| | - Minli Yu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (M.S.); (K.S.); (Q.Z.); (Y.D.); (Y.S.); (J.T.); (K.H.); (D.L.); (M.Y.)
| | - Yangqing Lu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China;
| | - Yanfei Tang
- Guangxi Fufeng Agricultural and Animal Husbandry Group Co., Ltd., Nanning 530024, China;
| | - Chungang Feng
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (M.S.); (K.S.); (Q.Z.); (Y.D.); (Y.S.); (J.T.); (K.H.); (D.L.); (M.Y.)
- Correspondence:
| |
Collapse
|
5
|
Wen Y, Dungan CM, Mobley CB, Valentino T, von Walden F, Murach KA. Nucleus Type-Specific DNA Methylomics Reveals Epigenetic "Memory" of Prior Adaptation in Skeletal Muscle. FUNCTION (OXFORD, ENGLAND) 2021; 2:zqab038. [PMID: 34870208 PMCID: PMC8636928 DOI: 10.1093/function/zqab038] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/28/2021] [Accepted: 08/03/2021] [Indexed: 02/07/2023]
Abstract
Using a mouse model of conditional and inducible in vivo fluorescent myonuclear labeling (HSA-GFP), sorting purification of nuclei, low-input reduced representation bisulfite sequencing (RRBS), and a translatable and reversible model of exercise (progressive weighted wheel running, PoWeR), we provide the first nucleus type-specific epigenetic information on skeletal muscle adaptation and detraining. Adult (>4 mo) HSA-GFP mice performed PoWeR for 8 wk then detrained for 12 wk; age-matched untrained mice were used to control for the long duration of the study. Myonuclei and interstitial nuclei from plantaris muscles were isolated for RRBS. Relative to untrained, PoWeR caused similar myonuclear CpG hypo- and hyper-methylation of promoter regions and substantial hypomethylation in interstitial nuclear promoters. Over-representation analysis of promoters revealed a larger number of hyper- versus hypo-methylated pathways in both nuclear populations after training and evidence for reciprocal regulation of methylation between nucleus types, with hypomethylation of promoter regions in Wnt signaling-related genes in myonuclei and hypermethylation in interstitial nuclei. After 12 wk of detraining, promoter CpGs in documented muscle remodeling-associated genes and pathways that were differentially methylated immediately after PoWeR were persistently differentially methylated in myonuclei, along with long-term promoter hypomethylation in interstitial nuclei. No enduring gene expression changes in muscle tissue were observed using RNA-sequencing. Upon 4 wk of retraining, mice that trained previously grew more at the whole muscle and fiber type-specific cellular level than training naïve mice, with no difference in myonuclear number. Muscle nuclei have a methylation epi-memory of prior training that may augment muscle adaptability to retraining.
Collapse
Affiliation(s)
- Yuan Wen
- Department of Physiology, University of Kentucky, Lexington, KY 40508, USA,The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Cory M Dungan
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA,College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - C Brooks Mobley
- Department of Physiology, University of Kentucky, Lexington, KY 40508, USA,The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Taylor Valentino
- Department of Physiology, University of Kentucky, Lexington, KY 40508, USA,The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Ferdinand von Walden
- Division of Pediatric Neurology, Department of Women's and Children's Health, Karolinska Institutet, Stockholm 171 77, Sweden
| | | |
Collapse
|
6
|
Xia Q, Ling X, Wang Z, Shen T, Chen M, Mao D, Ma X, Ning J, Zhang H, Chen D, Gu Q, Shen H, Yan J. Lateral rectus muscle differentiation potential in paralytic esotropia patients. BMC Ophthalmol 2021; 21:235. [PMID: 34044792 PMCID: PMC8161593 DOI: 10.1186/s12886-021-01994-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 05/10/2021] [Indexed: 12/05/2022] Open
Abstract
Purpose and background Recently, we found that maximal medial rectus recession and lateral rectus resection in patients with complete lateral rectus paralysis resulted in a partial restoration of abduction. In an attempt to understand some of the mechanisms involved with this effect we examined gene expression profiles of lateral recti from these patients, with our focus being directed to genes related to myogenesis. Materials and methods Lateral recti resected from patients with complete lateral rectus paralysis and those from concomitant esotropia (controls) were collected. Differences in gene expression profiles between these two groups were examined using microarray analysis and quantitative Reverse-transcription PCR (qRT-PCR). Results A total of 3056 differentially expressed genes (DEGs) were identified between these two groups. Within the paralytic esotropia group, 2081 genes were up-regulated and 975 down-regulated. The results of RT-PCR revealed that PAX7, MYOG, PITX1, SIX1 and SIX4 showed higher levels of expression, while that of MYOD a lower level of expression within the paralytic esotropia group as compared with that in the control group (p < 0.05). Conclusion The decreased expression of MYOD in the paralytic esotropia group suggested that extraocular muscle satellite cell (EOMSCs) differentiation processes were inhibited. Whereas the high expression levels of PAX7, SIX1/4 and MYOG, suggested that the EOMSCs were showing an effective potential for differentiation. The stimulation resulting from muscle surgery may induce EOMSCs to differentiate and thus restore abduction function. Supplementary Information The online version contains supplementary material available at 10.1186/s12886-021-01994-4.
Collapse
Affiliation(s)
- Qing Xia
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54 Xianlie Road, Guangzhou, 510060, China
| | - Xiangtian Ling
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54 Xianlie Road, Guangzhou, 510060, China
| | - Zhonghao Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54 Xianlie Road, Guangzhou, 510060, China
| | - Tao Shen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54 Xianlie Road, Guangzhou, 510060, China
| | - Minghao Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54 Xianlie Road, Guangzhou, 510060, China
| | - Danyi Mao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54 Xianlie Road, Guangzhou, 510060, China
| | - Xinqi Ma
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54 Xianlie Road, Guangzhou, 510060, China
| | - Jie Ning
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54 Xianlie Road, Guangzhou, 510060, China
| | - Han Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54 Xianlie Road, Guangzhou, 510060, China
| | - Dongli Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54 Xianlie Road, Guangzhou, 510060, China
| | - Qiong Gu
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, People's Republic of China
| | - Huangxuan Shen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54 Xianlie Road, Guangzhou, 510060, China. .,Biobank of Eye, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54 Xianlie Road, Guangzhou, 510060, China.
| | - Jianhua Yan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54 Xianlie Road, Guangzhou, 510060, China.
| |
Collapse
|
7
|
Karpukhina A, Galkin I, Ma Y, Dib C, Zinovkin R, Pletjushkina O, Chernyak B, Popova E, Vassetzky Y. Analysis of genes regulated by DUX4 via oxidative stress reveals potential therapeutic targets for treatment of facioscapulohumeral dystrophy. Redox Biol 2021; 43:102008. [PMID: 34030118 PMCID: PMC8163973 DOI: 10.1016/j.redox.2021.102008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 05/04/2021] [Accepted: 05/09/2021] [Indexed: 12/27/2022] Open
Abstract
Muscles of patients with facioscapulohumeral dystrophy (FSHD) are characterized by sporadic DUX4 expression and oxidative stress which is at least partially induced by DUX4 protein. Nevertheless, targeting oxidative stress with antioxidants has a limited impact on FSHD patients, and the exact role of oxidative stress in the pathology of FSHD, as well as its interplay with the DUX4 expression, remain unclear. Here we set up a screen for genes that are upregulated by DUX4 via oxidative stress with the aim to target these genes rather than the oxidative stress itself. Immortalized human myoblasts expressing DUX4 (MB135-DUX4) have an increased level of reactive oxygen species (ROS) and exhibit differentiation defects which can be reduced by treating the cells with classic (Tempol) or mitochondria-targeted antioxidants (SkQ1). The transcriptome analysis of antioxidant-treated MB135 and MB135-DUX4 myoblasts allowed us to identify 200 genes with expression deregulated by DUX4 but normalized upon antioxidant treatment. Several of these genes, including PITX1, have been already associated with FSHD and/or muscle differentiation. We confirmed that PITX1 was indeed deregulated in MB135-DUX4 cells and primary FSHD myoblasts and revealed a redox component in PITX1 regulation. PITX1 silencing partially reversed the differentiation defects of MB135-DUX4 myoblasts. Our approach can be used to identify and target redox-dependent genes involved in human diseases. Double homeobox transcription factor DUX4 misregulates hundreds of genes and induces oxidative stress in human myoblasts. ROS, notably those of mitochondrial origin, contribute to the differentiation defects in myoblasts expressing DUX4. A subset of genes is deregulated by DUX4 indirectly, via oxidative stress. A strategy to identify the genes deregulated by DUX4 via oxidative stress was developed. PITX1 is deregulated by DUX4 via oxidative stress and can be targeted to improve myogenesis in DUX4-expressing myoblasts.
Collapse
Affiliation(s)
- Anna Karpukhina
- CNRS UMR9018, Université Paris-Saclay, Institut Gustave Roussy, 94805, Villejuif, France; Koltzov Institute of Developmental Biology, 117334, Moscow, Russia; Faculty of Bioengineering and Bioinformatics, MSU, 119992, Moscow, Russia
| | - Ivan Galkin
- Belozersky Institute of Physico-Chemical Biology, 119992, Moscow, Russia
| | - Yinxing Ma
- CNRS UMR9018, Université Paris-Saclay, Institut Gustave Roussy, 94805, Villejuif, France
| | - Carla Dib
- CNRS UMR9018, Université Paris-Saclay, Institut Gustave Roussy, 94805, Villejuif, France
| | - Roman Zinovkin
- Belozersky Institute of Physico-Chemical Biology, 119992, Moscow, Russia
| | - Olga Pletjushkina
- Belozersky Institute of Physico-Chemical Biology, 119992, Moscow, Russia
| | - Boris Chernyak
- Belozersky Institute of Physico-Chemical Biology, 119992, Moscow, Russia
| | - Ekaterina Popova
- Belozersky Institute of Physico-Chemical Biology, 119992, Moscow, Russia
| | - Yegor Vassetzky
- CNRS UMR9018, Université Paris-Saclay, Institut Gustave Roussy, 94805, Villejuif, France; Koltzov Institute of Developmental Biology, 117334, Moscow, Russia.
| |
Collapse
|
8
|
Schätzl T, Kaiser L, Deigner HP. Facioscapulohumeral muscular dystrophy: genetics, gene activation and downstream signalling with regard to recent therapeutic approaches: an update. Orphanet J Rare Dis 2021; 16:129. [PMID: 33712050 PMCID: PMC7953708 DOI: 10.1186/s13023-021-01760-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 02/25/2021] [Indexed: 12/12/2022] Open
Abstract
Whilst a disease-modifying treatment for Facioscapulohumeral muscular dystrophy (FSHD) does not exist currently, recent advances in complex molecular pathophysiology studies of FSHD have led to possible therapeutic approaches for its targeted treatment. Although the underlying genetics of FSHD have been researched extensively, there remains an incomplete understanding of the pathophysiology of FSHD in relation to the molecules leading to DUX4 gene activation and the downstream gene targets of DUX4 that cause its toxic effects. In the context of the local proximity of chromosome 4q to the nuclear envelope, a contraction of the D4Z4 macrosatellite induces lower methylation levels, enabling the ectopic expression of DUX4. This disrupts numerous signalling pathways that mostly result in cell death, detrimentally affecting skeletal muscle in affected individuals. In this regard different options are currently explored either to suppress the transcription of DUX4 gene, inhibiting DUX4 protein from its toxic effects, or to alleviate the symptoms triggered by its numerous targets.
Collapse
Affiliation(s)
- Teresa Schätzl
- Institute of Precision Medicine, Medical and Life Sciences Faculty, Furtwangen University, Jakob-Kienzle-Straße 17, 78054, Villingen-Schwenningen, Germany
| | - Lars Kaiser
- Institute of Precision Medicine, Medical and Life Sciences Faculty, Furtwangen University, Jakob-Kienzle-Straße 17, 78054, Villingen-Schwenningen, Germany
- Institute of Pharmaceutical Sciences, University of Freiburg, Albertstraße 25, 79104, Freiburg i. Br., Germany
| | - Hans-Peter Deigner
- Institute of Precision Medicine, Medical and Life Sciences Faculty, Furtwangen University, Jakob-Kienzle-Straße 17, 78054, Villingen-Schwenningen, Germany.
- EXIM Department, Fraunhofer Institute IZI, Leipzig, Schillingallee 68, 18057, Rostock, Germany.
- Faculty of Science, Tuebingen University, Auf der Morgenstelle 8, 72076, Tübingen, Germany.
| |
Collapse
|
9
|
Lim KRQ, Yokota T. Genetic Approaches for the Treatment of Facioscapulohumeral Muscular Dystrophy. Front Pharmacol 2021; 12:642858. [PMID: 33776777 PMCID: PMC7996372 DOI: 10.3389/fphar.2021.642858] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/01/2021] [Indexed: 12/26/2022] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is an autosomal dominant disorder characterized by progressive, asymmetric muscle weakness at the face, shoulders, and upper limbs, which spreads to the lower body with age. It is the third most common inherited muscular disorder worldwide. Around 20% of patients are wheelchair-bound, and some present with extramuscular manifestations. FSHD is caused by aberrant expression of the double homeobox protein 4 (DUX4) gene in muscle. DUX4 codes for a transcription factor which, in skeletal muscle, dysregulates numerous signaling activities that culminate in cytotoxicity. Potential treatments for FSHD therefore aim to reduce the expression of DUX4 or the activity of its toxic protein product. In this article, we review how genetic approaches such as those based on oligonucleotide and genome editing technologies have been developed to achieve these goals. We also outline the challenges these therapies are facing on the road to translation, and discuss possible solutions and future directions.
Collapse
Affiliation(s)
- Kenji Rowel Q. Lim
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Toshifumi Yokota
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- The Friends of Garrett Cumming Research and Muscular Dystrophy Canada, HM Toupin Neurological Science Research Chair, Edmonton, AB, Canada
| |
Collapse
|
10
|
Lim KRQ, Bittel A, Maruyama R, Echigoya Y, Nguyen Q, Huang Y, Dzierlega K, Zhang A, Chen YW, Yokota T. DUX4 Transcript Knockdown with Antisense 2'-O-Methoxyethyl Gapmers for the Treatment of Facioscapulohumeral Muscular Dystrophy. Mol Ther 2021; 29:848-858. [PMID: 33068777 PMCID: PMC7854280 DOI: 10.1016/j.ymthe.2020.10.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 09/24/2020] [Accepted: 10/12/2020] [Indexed: 01/11/2023] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is an autosomal dominant disorder characterized by a progressive, asymmetric weakening of muscles, starting with those in the upper body. It is caused by aberrant expression of the double homeobox protein 4 gene (DUX4) in skeletal muscle. FSHD is currently incurable. We propose to develop a therapy for FSHD using antisense 2'-O-methoxyethyl (2'-MOE) gapmers, to knock down DUX4 mRNA expression. Using immortalized patient-derived muscle cells and local intramuscular injections in the FLExDUX4 FSHD mouse model, we showed that our designed 2'-MOE gapmers significantly reduced DUX4 transcript levels in vitro and in vivo, respectively. Furthermore, in vitro, we observed significantly reduced expression of DUX4-activated downstream targets, restoration of FSHD signature genes by RNA sequencing, significant improvements in myotube morphology, and minimal off-target activity. This work facilitates the development of a promising candidate therapy for FSHD and lays down the foundation for in vivo systemic treatment studies.
Collapse
Affiliation(s)
- Kenji Rowel Q Lim
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2H7, Canada
| | - Adam Bittel
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC 20010, USA
| | - Rika Maruyama
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2H7, Canada
| | - Yusuke Echigoya
- Laboratory of Biomedical Science, Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, Fujisawa, Kanagawa 252-0880, Japan
| | - Quynh Nguyen
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2H7, Canada
| | - Yiqing Huang
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2H7, Canada
| | - Kasia Dzierlega
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2H7, Canada
| | - Aiping Zhang
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC 20010, USA
| | - Yi-Wen Chen
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC 20010, USA; Department of Genomics and Precision Medicine, School of Medicine and Health Science, George Washington University, Washington, DC 20052, USA.
| | - Toshifumi Yokota
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2H7, Canada; Muscular Dystrophy Canada Research Chair, Edmonton, AB T6G2H7, Canada.
| |
Collapse
|
11
|
Chiu W, Hsun YH, Chang KJ, Yarmishyn AA, Hsiao YJ, Chien Y, Chien CS, Ma C, Yang YP, Tsai PH, Chiou SH, Lin TY, Cheng HM. Current Genetic Survey and Potential Gene-Targeting Therapeutics for Neuromuscular Diseases. Int J Mol Sci 2020; 21:E9589. [PMID: 33339321 PMCID: PMC7767109 DOI: 10.3390/ijms21249589] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/08/2020] [Accepted: 12/14/2020] [Indexed: 12/17/2022] Open
Abstract
Neuromuscular diseases (NMDs) belong to a class of functional impairments that cause dysfunctions of the motor neuron-muscle functional axis components. Inherited monogenic neuromuscular disorders encompass both muscular dystrophies and motor neuron diseases. Understanding of their causative genetic defects and pathological genetic mechanisms has led to the unprecedented clinical translation of genetic therapies. Challenged by a broad range of gene defect types, researchers have developed different approaches to tackle mutations by hijacking the cellular gene expression machinery to minimize the mutational damage and produce the functional target proteins. Such manipulations may be directed to any point of the gene expression axis, such as classical gene augmentation, modulating premature termination codon ribosomal bypass, splicing modification of pre-mRNA, etc. With the soar of the CRISPR-based gene editing systems, researchers now gravitate toward genome surgery in tackling NMDs by directly correcting the mutational defects at the genome level and expanding the scope of targetable NMDs. In this article, we will review the current development of gene therapy and focus on NMDs that are available in published reports, including Duchenne Muscular Dystrophy (DMD), Becker muscular dystrophy (BMD), X-linked myotubular myopathy (XLMTM), Spinal Muscular Atrophy (SMA), and Limb-girdle muscular dystrophy Type 2C (LGMD2C).
Collapse
Affiliation(s)
- Wei Chiu
- Department of Medicine, National Yang-Ming University, Taipei 11221, Taiwan; (W.C.); (K.-J.C.); (Y.-J.H.); (Y.C.); (Y.-P.Y.); (S.-H.C.)
| | - Ya-Hsin Hsun
- Department of Psychology, University of Toronto, Toronto, ON M1C 1A4, Canada;
- Department of Biological Science, University of Toronto, Toronto, ON M1C 1A4, Canada
| | - Kao-Jung Chang
- Department of Medicine, National Yang-Ming University, Taipei 11221, Taiwan; (W.C.); (K.-J.C.); (Y.-J.H.); (Y.C.); (Y.-P.Y.); (S.-H.C.)
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 11221, Taiwan
| | - Aliaksandr A. Yarmishyn
- Division of Basic Research, Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan; (A.A.Y.); (P.-H.T.)
| | - Yu-Jer Hsiao
- Department of Medicine, National Yang-Ming University, Taipei 11221, Taiwan; (W.C.); (K.-J.C.); (Y.-J.H.); (Y.C.); (Y.-P.Y.); (S.-H.C.)
| | - Yueh Chien
- Department of Medicine, National Yang-Ming University, Taipei 11221, Taiwan; (W.C.); (K.-J.C.); (Y.-J.H.); (Y.C.); (Y.-P.Y.); (S.-H.C.)
- Division of Basic Research, Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan; (A.A.Y.); (P.-H.T.)
| | - Chian-Shiu Chien
- Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei 112201, Taiwan;
- Institute of Pharmacology, National Yang-Ming University, Taipei 11221, Taiwan
| | - Chun Ma
- Department of Medicine, National Taiwan University, Taipei 10617, Taiwan;
| | - Yi-Ping Yang
- Department of Medicine, National Yang-Ming University, Taipei 11221, Taiwan; (W.C.); (K.-J.C.); (Y.-J.H.); (Y.C.); (Y.-P.Y.); (S.-H.C.)
- Division of Basic Research, Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan; (A.A.Y.); (P.-H.T.)
- Institute of Food Safety and Health Risk Assessment, National Yang-Ming University, Taipei 11221, Taiwan
| | - Ping-Hsing Tsai
- Division of Basic Research, Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan; (A.A.Y.); (P.-H.T.)
- Institute of Pharmacology, National Yang-Ming University, Taipei 11221, Taiwan
| | - Shih-Hwa Chiou
- Department of Medicine, National Yang-Ming University, Taipei 11221, Taiwan; (W.C.); (K.-J.C.); (Y.-J.H.); (Y.C.); (Y.-P.Y.); (S.-H.C.)
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 11221, Taiwan
- Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei 112201, Taiwan;
- Institute of Pharmacology, National Yang-Ming University, Taipei 11221, Taiwan
- Institute of Food Safety and Health Risk Assessment, National Yang-Ming University, Taipei 11221, Taiwan
- Genomic Research Center, Academia Sinica, Taipei 11529, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Chiao-Tung University, Hsinchu 1001, Taiwan
| | - Ting-Yi Lin
- Department of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Hao-Min Cheng
- Department of Medicine, National Yang-Ming University, Taipei 11221, Taiwan; (W.C.); (K.-J.C.); (Y.-J.H.); (Y.C.); (Y.-P.Y.); (S.-H.C.)
- Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei 112201, Taiwan;
- Center for Evidence-based Medicine, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| |
Collapse
|
12
|
Heier CR, Zhang A, Nguyen NY, Tully CB, Panigrahi A, Gordish-Dressman H, Pandey SN, Guglieri M, Ryan MM, Clemens PR, Thangarajh M, Webster R, Smith EC, Connolly AM, McDonald CM, Karachunski P, Tulinius M, Harper A, Mah JK, Fiorillo AA, Chen YW. Multi-Omics Identifies Circulating miRNA and Protein Biomarkers for Facioscapulohumeral Dystrophy. J Pers Med 2020; 10:jpm10040236. [PMID: 33228131 PMCID: PMC7711540 DOI: 10.3390/jpm10040236] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/04/2020] [Accepted: 11/06/2020] [Indexed: 12/15/2022] Open
Abstract
The development of therapeutics for muscle diseases such as facioscapulohumeral dystrophy (FSHD) is impeded by a lack of objective, minimally invasive biomarkers. Here we identify circulating miRNAs and proteins that are dysregulated in early-onset FSHD patients to develop blood-based molecular biomarkers. Plasma samples from clinically characterized individuals with early-onset FSHD provide a discovery group and are compared to healthy control volunteers. Low-density quantitative polymerase chain reaction (PCR)-based arrays identify 19 candidate miRNAs, while mass spectrometry proteomic analysis identifies 13 candidate proteins. Bioinformatic analysis of chromatin immunoprecipitation (ChIP)-seq data shows that the FSHD-dysregulated DUX4 transcription factor binds to regulatory regions of several candidate miRNAs. This panel of miRNAs also shows ChIP signatures consistent with regulation by additional transcription factors which are up-regulated in FSHD (FOS, EGR1, MYC, and YY1). Validation studies in a separate group of patients with FSHD show consistent up-regulation of miR-100, miR-103, miR-146b, miR-29b, miR-34a, miR-454, miR-505, and miR-576. An increase in the expression of S100A8 protein, an inflammatory regulatory factor and subunit of calprotectin, is validated by Enzyme-Linked Immunosorbent Assay (ELISA). Bioinformatic analyses of proteomics and miRNA data further support a model of calprotectin and toll-like receptor 4 (TLR4) pathway dysregulation in FSHD. Moving forward, this panel of miRNAs, along with S100A8 and calprotectin, merit further investigation as monitoring and pharmacodynamic biomarkers for FSHD.
Collapse
Affiliation(s)
- Christopher R. Heier
- Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA; (H.G.-D.); (A.A.F.)
- Correspondence: (C.R.H.); (Y.-W.C.)
| | - Aiping Zhang
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC 20010, USA; (A.Z.); (N.Y.N.); (C.B.T.); (A.P.); (S.N.P.)
| | - Nhu Y Nguyen
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC 20010, USA; (A.Z.); (N.Y.N.); (C.B.T.); (A.P.); (S.N.P.)
| | - Christopher B. Tully
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC 20010, USA; (A.Z.); (N.Y.N.); (C.B.T.); (A.P.); (S.N.P.)
| | - Aswini Panigrahi
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC 20010, USA; (A.Z.); (N.Y.N.); (C.B.T.); (A.P.); (S.N.P.)
| | - Heather Gordish-Dressman
- Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA; (H.G.-D.); (A.A.F.)
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC 20010, USA; (A.Z.); (N.Y.N.); (C.B.T.); (A.P.); (S.N.P.)
| | - Sachchida Nand Pandey
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC 20010, USA; (A.Z.); (N.Y.N.); (C.B.T.); (A.P.); (S.N.P.)
| | | | - Monique M. Ryan
- The Royal Children’s Hospital, Melbourne University, Parkville, Victoria 3052, Australia;
| | - Paula R. Clemens
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA;
| | - Mathula Thangarajh
- Department of Neurology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA;
| | | | - Edward C. Smith
- Department of Pediatrics, Duke University Medical Center, Durham, NC 27705, USA;
| | - Anne M. Connolly
- Nationwide Children’s Hospital, The Ohio State University, Columbus, OH 43205, USA;
| | - Craig M. McDonald
- Department of Physical Medicine and Rehabilitation, University of California at Davis Medical Center, Sacramento, CA 95817, USA;
| | - Peter Karachunski
- Department of Neurology, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Mar Tulinius
- Department of Pediatrics, Gothenburg University, Queen Silvia Children’s Hospital, 41685 Göteborg, Sweden;
| | - Amy Harper
- Department of Neurology, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Jean K. Mah
- Deparment of Pediatrics and Clinical Neurosciences, Cumming School of Medicine, University of Calgary, T2N T3B, Calgary, AB 6A81N4, Canada;
| | - Alyson A. Fiorillo
- Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA; (H.G.-D.); (A.A.F.)
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC 20010, USA; (A.Z.); (N.Y.N.); (C.B.T.); (A.P.); (S.N.P.)
| | - Yi-Wen Chen
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC 20010, USA; (A.Z.); (N.Y.N.); (C.B.T.); (A.P.); (S.N.P.)
- Correspondence: (C.R.H.); (Y.-W.C.)
| | | |
Collapse
|
13
|
Karpukhina A, Vassetzky Y. DUX4, a Zygotic Genome Activator, Is Involved in Oncogenesis and Genetic Diseases. Russ J Dev Biol 2020. [DOI: 10.1134/s1062360420030078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
14
|
Liu CW, Huang CC, Hsu CF, Li TH, Tsai YL, Lin MW, Tsai HC, Huang SF, Yang YY, Hsieh YC, Lee TY, Tsai CY, Huang YH, Hou MC, Lin HC. SIRT1-dependent mechanisms and effects of resveratrol for amelioration of muscle wasting in NASH mice. BMJ Open Gastroenterol 2020; 7:bmjgast-2020-000381. [PMID: 32371503 PMCID: PMC7228468 DOI: 10.1136/bmjgast-2020-000381] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/24/2020] [Accepted: 03/27/2020] [Indexed: 12/21/2022] Open
Abstract
Background In non-alcoholic steatohepatitis (NASH), muscle wasting was an aggravating factor for the progression of hepatic steatosis. This study explores the potential benefits of chronic treatment with resveratrol, a strong activator of SIRT1 on the muscle wasting of NASH mice. Methods In vivo and in vitro study, we evaluate the SIRT1-dependent mechanisms and effects of resveratrol administration for 6 weeks with high-fat-methionine and choline deficient diet-induced NASH mice and palmitate-pretreated C2C12 myoblast cells. Results Resveratrol treatment improved grip strength and muscle mass of limbs, increased running distance and time on exercise wheels in NASH mice. There is a negative correlation between muscular SIRT1 activity and 3-nitrotyrosine levels of NASH and NASH-resv mice. The SIRT1-dependent effect of muscle wasting was associated with the suppression of oxidative stress, upregulation of antioxidants, inhibition of protein degradation, activation of autophagy, suppression of apoptotic activity, upregulation of lipolytic genes and the reduction of fatty infiltration in limb muscles of NASH mice. In vitro, resveratrol alleviated palmitate acid-induced oxidative stress, lipid deposition, autophagy dysfunction, apoptotic signals, and subsequently reduced fusion index and myotube formation of C2C12 cells. The beneficial effects of resveratrol were abolished by EX527. Conclusions Our study suggests that chronic resveratrol treatment is a potential strategy for amelioration of hepatic steatosis and muscle wasting in NASH mouse model.
Collapse
Affiliation(s)
- Chih-Wei Liu
- Division of Allergy, Immunology and Rheumatology, Taipei, Taiwan.,Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute of Clinical Medicine, Taipei, Taiwan
| | - Chia-Chang Huang
- Institute of Clinical Medicine, Taipei, Taiwan.,Division of Clinical Skills Center, Department of Medical Education, Taipei Veterans General Hospital, Taoyuan, Taiwan.,Faculty of Medicine, School of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Chien-Fu Hsu
- Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tzu-Hao Li
- Institute of Clinical Medicine, Taipei, Taiwan.,Faculty of Medicine, School of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan.,Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Yu-Lien Tsai
- Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ming-Wei Lin
- Faculty of Medicine, School of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan.,Institute of Public Health, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Hung-Cheng Tsai
- Division of Allergy, Immunology and Rheumatology, Taipei, Taiwan.,Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shiang-Fen Huang
- Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Faculty of Medicine, School of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan.,Division of Infection Disease, Taipei, Taiwan
| | - Ying-Ying Yang
- Institute of Clinical Medicine, Taipei, Taiwan .,Division of Clinical Skills Center, Department of Medical Education, Taipei Veterans General Hospital, Taoyuan, Taiwan.,Faculty of Medicine, School of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan.,Division of Gastroenterology and Hepatology, Taipei, Taiwan
| | - Yun-Cheng Hsieh
- Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Faculty of Medicine, School of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan.,Division of Gastroenterology and Hepatology, Taipei, Taiwan
| | - Tzung-Yan Lee
- Graduate Institute of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Chang-Youh Tsai
- Division of Allergy, Immunology and Rheumatology, Taipei, Taiwan.,Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Faculty of Medicine, School of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Yi-Hsiang Huang
- Institute of Clinical Medicine, Taipei, Taiwan.,Faculty of Medicine, School of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan.,Division of Gastroenterology and Hepatology, Taipei, Taiwan
| | - Ming-Chih Hou
- Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Faculty of Medicine, School of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan.,Division of Gastroenterology and Hepatology, Taipei, Taiwan
| | - Han-Chieh Lin
- Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Faculty of Medicine, School of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan.,Division of Gastroenterology and Hepatology, Taipei, Taiwan
| |
Collapse
|
15
|
Lim KRQ, Nguyen Q, Yokota T. DUX4 Signalling in the Pathogenesis of Facioscapulohumeral Muscular Dystrophy. Int J Mol Sci 2020; 21:E729. [PMID: 31979100 PMCID: PMC7037115 DOI: 10.3390/ijms21030729] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 01/17/2020] [Accepted: 01/18/2020] [Indexed: 12/17/2022] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is a disabling inherited muscular disorder characterized by asymmetric, progressive muscle weakness and degeneration. Patients display widely variable disease onset and severity, and sometimes present with extra-muscular symptoms. There is a consensus that FSHD is caused by the aberrant production of the double homeobox protein 4 (DUX4) transcription factor in skeletal muscle. DUX4 is normally expressed during early embryonic development, and is then effectively silenced in all tissues except the testis and thymus. Its reactivation in skeletal muscle disrupts numerous signalling pathways that mostly converge on cell death. Here, we review studies on DUX4-affected pathways in skeletal muscle and provide insights into how understanding these could help explain the unique pathogenesis of FSHD.
Collapse
Affiliation(s)
- Kenji Rowel Q. Lim
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2H7, Canada; (K.R.Q.L.); (Q.N.)
| | - Quynh Nguyen
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2H7, Canada; (K.R.Q.L.); (Q.N.)
| | - Toshifumi Yokota
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2H7, Canada; (K.R.Q.L.); (Q.N.)
- The Friends of Garrett Cumming Research & Muscular Dystrophy Canada, HM Toupin Neurological Science Research Chair, Edmonton, AB T6G2H7, Canada
| |
Collapse
|
16
|
Ding J, Li F, Cong Y, Miao J, Wu D, Liu B, Wang L. Trichostatin A inhibits skeletal muscle atrophy induced by cigarette smoke exposure in mice. Life Sci 2019; 235:116800. [PMID: 31472151 DOI: 10.1016/j.lfs.2019.116800] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 08/21/2019] [Accepted: 08/27/2019] [Indexed: 12/20/2022]
Abstract
AIMS It is well known that cigarette smoke (CS) is the main risk factor for chronic obstructive pulmonary disease (COPD) accompanied by skeletal muscle atrophy. Histone deacetylases (HDACs) that remove acetyl groups from target proteins are necessary for the muscle atrophy associated with skeletal muscle disuse. However, the role of HDACs and trichostatin A (TSA), a HDAC inhibitor, in skeletal muscle atrophy caused by CS exposure remains poorly understood. MAIN METHODS Female mice were exposed to CS twice daily for 40 days and TSA injected intraperitoneally into CS-exposed mice on alternate days. Skeletal muscles were weighed and gastrocnemius (Gas) muscle histomorphology examined by hematoxylin and eosin staining. Histone deacetylases 1 and 2 (HDAC1/2), and markers of ubiquitin degradation, muscle differentiation, apoptosis, pyroptosis, and the cytoskeletal proteins were assessed by western blot and immunohistochemistry in Gas. KEYFINDINGS CS exposure decreased body and skeletal muscle weights and triggered an increase in the percentage of fiber with centralized nuclei in Gas. HDAC1/2 proteins were upregulated in the Gas of mice exposed to CS, while TSA effectively inhibited HDAC1/2 protein levels and attenuated the loss of body weight and skeletal muscle wet weight induced by CS exposure. Markers for ubiquitin degradation, muscle differentiation, cytoskeletal proteins, apoptosis and pyroptosis were all upregulated following CS exposure and effectively restored by TSA. SIGNIFICANCE TSA may inhibit skeletal muscle atrophy and histomorphological alterations induced by CS exposure by downregulating markers of ubiquitin degradation, muscle fiber differentiation, cytoskeletal proteins, apoptosis and pyroptosis via HDAC1/2 inhibition.
Collapse
Affiliation(s)
- Jingjing Ding
- Medical Research Center of Shengjing Hospital, China Medical University, Shenyang 110004, China; Key Laboratory of Research and Application of Animal Model for Environmental and Metabolic Diseases, Liaoning Province, China
| | - Fang Li
- Medical Research Center of Shengjing Hospital, China Medical University, Shenyang 110004, China; Key Laboratory of Research and Application of Animal Model for Environmental and Metabolic Diseases, Liaoning Province, China
| | - Yanfei Cong
- Medical Research Center of Shengjing Hospital, China Medical University, Shenyang 110004, China; Key Laboratory of Research and Application of Animal Model for Environmental and Metabolic Diseases, Liaoning Province, China
| | - Jianing Miao
- Medical Research Center of Shengjing Hospital, China Medical University, Shenyang 110004, China; Key Laboratory of Research and Application of Animal Model for Environmental and Metabolic Diseases, Liaoning Province, China
| | - Di Wu
- Medical Research Center of Shengjing Hospital, China Medical University, Shenyang 110004, China; Key Laboratory of Research and Application of Animal Model for Environmental and Metabolic Diseases, Liaoning Province, China
| | - Bo Liu
- Medical Research Center of Shengjing Hospital, China Medical University, Shenyang 110004, China; Key Laboratory of Research and Application of Animal Model for Environmental and Metabolic Diseases, Liaoning Province, China
| | - Lili Wang
- Medical Research Center of Shengjing Hospital, China Medical University, Shenyang 110004, China; Key Laboratory of Research and Application of Animal Model for Environmental and Metabolic Diseases, Liaoning Province, China.
| |
Collapse
|
17
|
de Las Heras-Saldana S, Chung KY, Lee SH, Gondro C. Gene expression of Hanwoo satellite cell differentiation in longissimus dorsi and semimembranosus. BMC Genomics 2019; 20:156. [PMID: 30808286 PMCID: PMC6390542 DOI: 10.1186/s12864-019-5530-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 02/13/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Korean Hanwoo cattle are known for their high meat quality, especially their high intramuscular fat compared to most other cattle breeds. Different muscles have very different meat quality traits and a study of the myogenic process in satellite cells can help us better understand the genes and pathways that regulate this process and how muscles differentiate. RESULTS Cell cultures of Longissimus dorsi muscle differentiated from myoblast into multinucleated myotubes faster than semimembranosus. Time-series RNA-seq identified a total of 13 differentially expressed genes between the two muscles during their development. These genes seem to be involved in determining muscle lineage development and appear to modulate the expression of myogenic regulatory factors (mainly MYOD and MYF5) during differentiation of satellite cells into multinucleate myotubes. Gene ontology enriched terms were consistent with the morphological changes observed in the histology. Most of the over-represented terms and genes expressed during myoblast differentiation were similar regardless of muscle type which indicates a highly conserved myogenic process albeit the rates of differentiation being different. There were more differences in the enriched GO terms during the end of proliferation compared to myoblast differentiation. CONCLUSIONS The use of satellite cells from newborn Hanwoo calves appears to be a good model to study embryonic myogenesis in muscle. Our findings provide evidence that the differential expression of HOXB2, HOXB4, HOXB9, HOXC8, FOXD1, IGFN1, ZIC2, ZIC4, HOXA11, HOXC11, PITX1, SIM2 and TBX4 genes could be involved in the differentiation of Longissimus dorsi and Semimembranosus muscles. These genes seem to modulate the muscle fate of the satellite cells during myogenesis through a differential expression profile that also controls the expression of some myogenic regulatory factors (MYOD and MYF5). The number of differentially expressed genes across time was unsurprisingly large. In relation to the baseline day 0, there were 631, 155, 175, 519 and 586 DE genes in LD, while in SM we found 204, 0, 615, 761 and 1154 DE genes at days 1, 2, 4, 7 and 14 respectively.
Collapse
Affiliation(s)
| | - Ki Yong Chung
- Hanwoo Research Institute, National Institute of Animal Science, RDA, Pyeongchang, South Korea
| | - Seung Hwan Lee
- Division of Animal and Dairy Science, Chungnam National University, Deajeon, South Korea.
| | - Cedric Gondro
- Department of Animal Science, Michigan State University, 474 S Shaw Lane, East Lansing, MI, 48824, USA.
| |
Collapse
|
18
|
Erekat N, Al-Jarrah MD. Interleukin-1 Beta and Tumor Necrosis Factor Alpha Upregulation and Nuclear Factor Kappa B Activation in Skeletal Muscle from a Mouse Model of Chronic/Progressive Parkinson Disease. Med Sci Monit 2018; 24:7524-7531. [PMID: 30344306 PMCID: PMC6402272 DOI: 10.12659/msm.909032] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Skeletal muscle atrophy has been reported in patients with Parkinson disease (PD). The purpose of this study was to examine the potential implication of interleukin 1 beta (IL-1β), tumor necrosis factor alpha (TNFα), and nuclear factor kappa B (NF kappa B) in skeletal muscle atrophy following PD induction. MATERIAL AND METHODS Chronic Parkinsonism was induced in 10 albino mice by MPTP/probenecid treatment, while 10 other albino mice remained without treatment and were subsequently used as controls. Gastrocnemius muscles were examined for the expression of IL-1β and TNF-α, as well as the nuclear localization of NF kappa B, indicative of its activation, using immunohistochemistry in the 2 different groups. RESULTS IL-1β and TNF-α expression and NF kappa B nuclear localization were significantly higher in the PD skeletal muscle compared with those in the controls (P value <0.01). CONCLUSIONS The present data are indicative of an association of PD with IL-1β and TNF-α upregulation and NF kappa B activation in gastrocnemius muscles, potentially promoting the atrophy frequently observed in PD.
Collapse
Affiliation(s)
- Nour Erekat
- Department of Anatomy, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Muhammed D Al-Jarrah
- Department of Rehabilitation Sciences, Faculty of Applied Medical Sciences, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
19
|
Shi B, Xu L, Mao S, Xu L, Liu Z, Sun X, Zhu Z, Qiu Y. Abnormal PITX1 gene methylation in adolescent idiopathic scoliosis: a pilot study. BMC Musculoskelet Disord 2018; 19:138. [PMID: 29743058 PMCID: PMC5941792 DOI: 10.1186/s12891-018-2054-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 04/23/2018] [Indexed: 01/18/2023] Open
Abstract
Background The gene of pituitary homeobox 1 (PITX1) has been reported to be down-regulated in adolescent idiopathic scoliosis (AIS), of which the cause has not been well addressed. The abnormal DNA methylation was recently assumed to be an important mechanism for the down-regulated genes expression. However, the association between PITX1 promoter methylation and the etiology of AIS was not clear. Methods The peripheral blood samples of 50 AIS patients and 50 healthy controls were collected and the genomic DNA was extracted. The pyrosequencing assay was used to assess the methylation status of PITX1 promoter and real-time quantitative polymerase chain reaction (PCR) was used to detect the PITX1 gene expression. Comparison analysis was performed using independent t test and Chi-square tests, while correlation analysis were performed with 2-tailed Pearson coefficients. Results The mean methylation level was (3.52 ± 0.96)% in AIS and (1.40 ± 0.81)% in healthy controls (P < 0.0001). The PITX1 gene expression was 0.15 ± 0.08 in AIS and 0.80 ± 0.55 in healthy controls (P < 0.0001). The comparative analysis showed significant difference in age (P = 0.021) and Cobb angle of the main curve (P = 0.0001) between AIS groups with positive and negative methylation. The methylation level of 6 CpG sites in PITX1 promoters was significantly associated with Cobb angle of the main curve (P < 0.001) in AIS. No statistical relationship between PITX1 promoter methylation and gene expression was found in AIS (P = 0.842). Conclusion Significantly higher methylation level and lower PITX1 gene expression are found in AIS patients. PITX1 methylation is associated with Cobb angles of the main curves in AIS. DNA methylation thus plays an important role in the etiology and curve progression in AIS.
Collapse
Affiliation(s)
- Benlong Shi
- Spine Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Zhongshan Road No. 321, Nanjing, 210008, China
| | - Liang Xu
- Spine Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Zhongshan Road No. 321, Nanjing, 210008, China
| | - Saihu Mao
- Spine Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Zhongshan Road No. 321, Nanjing, 210008, China
| | - Leilei Xu
- Spine Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Zhongshan Road No. 321, Nanjing, 210008, China
| | - Zhen Liu
- Spine Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Zhongshan Road No. 321, Nanjing, 210008, China
| | - Xu Sun
- Spine Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Zhongshan Road No. 321, Nanjing, 210008, China
| | - Zezhang Zhu
- Spine Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Zhongshan Road No. 321, Nanjing, 210008, China.
| | - Yong Qiu
- Spine Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Zhongshan Road No. 321, Nanjing, 210008, China
| |
Collapse
|
20
|
Jung HJ, Lee KP, Milholland B, Shin YJ, Kang JS, Kwon KS, Suh Y. Comprehensive miRNA Profiling of Skeletal Muscle and Serum in Induced and Normal Mouse Muscle Atrophy During Aging. J Gerontol A Biol Sci Med Sci 2017; 72:1483-1491. [PMID: 28329037 DOI: 10.1093/gerona/glx025] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 02/01/2017] [Indexed: 01/29/2023] Open
Abstract
Age-associated loss of muscle mass and function is a major cause of morbidity and mortality in the elderly adults. Muscular atrophy can also be induced by disuse associated with long-term bed rest or disease. Although miRNAs regulate muscle growth, regeneration, and aging, their potential role in acute muscle atrophy is poorly understood. Furthermore, alterations in circulating miRNA levels have been shown to occur during aging but their potential as noninvasive biomarkers for muscle atrophy remains largely unexplored. Here, we report comprehensive miRNA expression profiles by miRNA-seq analysis in tibialis anterior muscle and serum of a disuse-induced atrophy mouse model, mimicking the acute atrophy following long-term bed rest, as compared to those of young and old mice. Comparative analysis and validation studies have revealed that miR-455-3p was significantly decreased in muscle of both induced-atrophy model and old mice, whereas miR-434-3p was decreased in both serum and muscle of old mice, as compared to young mice. Furthermore, upregulation of miR-455-3p in fully differentiated C2C12 myoblasts induced a hypertrophic phenotype. These results suggest that deregulation of miR-455-3p may play a functional role in muscle atrophy and miR-434-3p could be a candidate serum biomarker of muscle aging.
Collapse
Affiliation(s)
- Hwa Jin Jung
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York
| | - Kwang-Pyo Lee
- Aging Research Center, Korea Research Institute of Bioscience & Biotechnology, Daejeon.,Department of Biomolecular Science
| | - Brandon Milholland
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York
| | - Yeo Jin Shin
- Aging Research Center, Korea Research Institute of Bioscience & Biotechnology, Daejeon.,Department of Functional Genomics, Korea University of Science and Technology, Daejeon
| | - Jae Sook Kang
- Aging Research Center, Korea Research Institute of Bioscience & Biotechnology, Daejeon.,Department of Functional Genomics, Korea University of Science and Technology, Daejeon
| | - Ki-Sun Kwon
- Aging Research Center, Korea Research Institute of Bioscience & Biotechnology, Daejeon.,Department of Functional Genomics, Korea University of Science and Technology, Daejeon
| | - Yousin Suh
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
21
|
DeSimone AM, Pakula A, Lek A, Emerson CP. Facioscapulohumeral Muscular Dystrophy. Compr Physiol 2017; 7:1229-1279. [PMID: 28915324 DOI: 10.1002/cphy.c160039] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Facioscapulohumeral Muscular Dystrophy is a common form of muscular dystrophy that presents clinically with progressive weakness of the facial, scapular, and humeral muscles, with later involvement of the trunk and lower extremities. While typically inherited as autosomal dominant, facioscapulohumeral muscular dystrophy (FSHD) has a complex genetic and epigenetic etiology that has only recently been well described. The most prevalent form of the disease, FSHD1, is associated with the contraction of the D4Z4 microsatellite repeat array located on a permissive 4qA chromosome. D4Z4 contraction allows epigenetic derepression of the array, and possibly the surrounding 4q35 region, allowing misexpression of the toxic DUX4 transcription factor encoded within the terminal D4Z4 repeat in skeletal muscles. The less common form of the disease, FSHD2, results from haploinsufficiency of the SMCHD1 gene in individuals carrying a permissive 4qA allele, also leading to the derepression of DUX4, further supporting a central role for DUX4. How DUX4 misexpression contributes to FSHD muscle pathology is a major focus of current investigation. Misexpression of other genes at the 4q35 locus, including FRG1 and FAT1, and unlinked genes, such as SMCHD1, has also been implicated as disease modifiers, leading to several competing disease models. In this review, we describe recent advances in understanding the pathophysiology of FSHD, including the application of MRI as a research and diagnostic tool, the genetic and epigenetic disruptions associated with the disease, and the molecular basis of FSHD. We discuss how these advances are leading to the emergence of new approaches to enable development of FSHD therapeutics. © 2017 American Physiological Society. Compr Physiol 7:1229-1279, 2017.
Collapse
Affiliation(s)
- Alec M DeSimone
- Wellstone Muscular Dystrophy Program, Department of Neurology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Anna Pakula
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Pediatrics and Genetics at Harvard Medical School, Boston, Massachusetts, USA
| | - Angela Lek
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Pediatrics and Genetics at Harvard Medical School, Boston, Massachusetts, USA.,Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Charles P Emerson
- Wellstone Muscular Dystrophy Program, Department of Neurology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
22
|
Denny AP, Heather AK. Are Antioxidants a Potential Therapy for FSHD? A Review of the Literature. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:7020295. [PMID: 28690764 PMCID: PMC5485364 DOI: 10.1155/2017/7020295] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 04/27/2017] [Accepted: 05/03/2017] [Indexed: 11/21/2022]
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is an inherited myopathy affecting approximately 1 in 7500 individuals worldwide. It is a progressive disease characterised by skeletal muscle weakness and wasting. A genetic mutation on the 4q35 chromosome results in the expression of the double homeobox 4 gene (DUX4) which drives oxidative stress, inflammation, toxicity, and atrophy within the skeletal muscle. FSHD is characterised by oxidative stress, and there is currently no cure and a lack of therapies for the disease. Antioxidants have been researched for many years, with investigators aiming to use antioxidants therapeutically for oxidative stress-associated diseases. This has included both natural and synthetic antioxidants. The use of antioxidants in preclinical or clinical models has been largely successful with a plethora of research reporting positive results. However, when translated to clinical trials, the use of antioxidants as a therapeutic intervention for a variety of disease has been largely unsuccessful. Moreover, specifically focusing on FSHD, limited research has been conducted on the use of antioxidants as a therapy in either preclinical or clinical models. This review summarises the current state of antioxidant use in the treatment of FSHD and discusses their potential avenue for therapeutic use for FSHD patients.
Collapse
Affiliation(s)
- Adam Philip Denny
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Alison Kay Heather
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
23
|
Teveroni E, Pellegrino M, Sacconi S, Calandra P, Cascino I, Farioli-Vecchioli S, Puma A, Garibaldi M, Morosetti R, Tasca G, Ricci E, Trevisan CP, Galluzzi G, Pontecorvi A, Crescenzi M, Deidda G, Moretti F. Estrogens enhance myoblast differentiation in facioscapulohumeral muscular dystrophy by antagonizing DUX4 activity. J Clin Invest 2017; 127:1531-1545. [PMID: 28263188 DOI: 10.1172/jci89401] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 01/12/2017] [Indexed: 01/28/2023] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is an autosomal dominant neuromuscular disorder that is characterized by extreme variability in symptoms, with females being less severely affected than males and presenting a higher proportion of asymptomatic carriers. The sex-related factors involved in the disease are not known. Here, we have utilized myoblasts isolated from FSHD patients (FSHD myoblasts) to investigate the effect of estrogens on muscle properties. Our results demonstrated that estrogens counteract the differentiation impairment of FSHD myoblasts without affecting cell proliferation or survival. Estrogen effects are mediated by estrogen receptor β (ERβ), which reduces chromatin occupancy and transcriptional activity of double homeobox 4 (DUX4), a protein whose aberrant expression has been implicated in FSHD pathogenesis. During myoblast differentiation, we observed that the levels and activity of DUX4 increased progressively and were associated with its enhanced recruitment in the nucleus. ERβ interfered with this recruitment by relocalizing DUX4 in the cytoplasm. This work identifies estrogens as a potential disease modifier that underlie sex-related differences in FSHD by protecting against myoblast differentiation impairments in this disease.
Collapse
|
24
|
Huffman KM, Jessee R, Andonian B, Davis BN, Narowski R, Huebner JL, Kraus VB, McCracken J, Gilmore BF, Tune KN, Campbell M, Koves TR, Muoio DM, Hubal MJ, Kraus WE. Molecular alterations in skeletal muscle in rheumatoid arthritis are related to disease activity, physical inactivity, and disability. Arthritis Res Ther 2017; 19:12. [PMID: 28114971 PMCID: PMC5260091 DOI: 10.1186/s13075-016-1215-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 12/30/2016] [Indexed: 01/04/2023] Open
Abstract
Background To identify molecular alterations in skeletal muscle in rheumatoid arthritis (RA) that may contribute to ongoing disability in RA. Methods Persons with seropositive or erosive RA (n = 51) and control subjects matched for age, gender, race, body mass index (BMI), and physical activity (n = 51) underwent assessment of disease activity, disability, pain, physical activity and thigh muscle biopsies. Muscle tissue was used for measurement of pro-inflammatory markers, transcriptomics, and comprehensive profiling of metabolic intermediates. Groups were compared using mixed models. Bivariate associations were assessed with Spearman correlation. Results Compared to controls, patients with RA had 75% greater muscle concentrations of IL-6 protein (p = 0.006). In patients with RA, muscle concentrations of inflammatory markers were positively associated (p < 0.05 for all) with disease activity (IL-1β, IL-8), disability (IL-1β, IL-6), pain (IL-1β, TNF-α, toll-like receptor (TLR)-4), and physical inactivity (IL-1β, IL-6). Muscle cytokines were not related to corresponding systemic cytokines. Prominent among the gene sets differentially expressed in muscles in RA versus controls were those involved in skeletal muscle repair processes and glycolytic metabolism. Metabolic profiling revealed 46% higher concentrations of pyruvate in muscle in RA (p < 0.05), and strong positive correlation between levels of amino acids involved in fibrosis (arginine, ornithine, proline, and glycine) and disability (p < 0.05). Conclusion RA is accompanied by broad-ranging molecular alterations in skeletal muscle. Analysis of inflammatory markers, gene expression, and metabolic intermediates linked disease-related disruptions in muscle inflammatory signaling, remodeling, and metabolic programming to physical inactivity and disability. Thus, skeletal muscle dysfunction might contribute to a viscous cycle of RA disease activity, physical inactivity, and disability. Electronic supplementary material The online version of this article (doi:10.1186/s13075-016-1215-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kim M Huffman
- Department of Medicine, Duke Molecular Physiology Institute, Duke School of Medicine, Durham, NC, USA.
| | - Ryan Jessee
- Department of Medicine, Duke Molecular Physiology Institute, Duke School of Medicine, Durham, NC, USA
| | - Brian Andonian
- Department of Medicine, Duke Molecular Physiology Institute, Duke School of Medicine, Durham, NC, USA
| | | | | | - Janet L Huebner
- Department of Medicine, Duke Molecular Physiology Institute, Duke School of Medicine, Durham, NC, USA
| | - Virginia B Kraus
- Department of Medicine, Duke Molecular Physiology Institute, Duke School of Medicine, Durham, NC, USA
| | - Julie McCracken
- Department of Medicine, Duke Molecular Physiology Institute, Duke School of Medicine, Durham, NC, USA
| | - Brian F Gilmore
- Department of Surgery, Duke School of Medicine, Durham, NC, USA
| | - K Noelle Tune
- Department of Emergency Medicine, Indiana University, Indianapolis, IN, USA
| | - Milton Campbell
- Department of Medicine, Duke Molecular Physiology Institute, Duke School of Medicine, Durham, NC, USA
| | - Timothy R Koves
- Department of Medicine, Duke Molecular Physiology Institute, Duke School of Medicine, Durham, NC, USA
| | - Deborah M Muoio
- Department of Medicine, Duke Molecular Physiology Institute, Duke School of Medicine, Durham, NC, USA
| | | | - William E Kraus
- Department of Medicine, Duke Molecular Physiology Institute, Duke School of Medicine, Durham, NC, USA
| |
Collapse
|
25
|
Caron L, Kher D, Lee KL, McKernan R, Dumevska B, Hidalgo A, Li J, Yang H, Main H, Ferri G, Petek LM, Poellinger L, Miller DG, Gabellini D, Schmidt U. A Human Pluripotent Stem Cell Model of Facioscapulohumeral Muscular Dystrophy-Affected Skeletal Muscles. Stem Cells Transl Med 2016; 5:1145-61. [PMID: 27217344 PMCID: PMC4996435 DOI: 10.5966/sctm.2015-0224] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 04/07/2016] [Indexed: 01/13/2023] Open
Abstract
UNLABELLED : Facioscapulohumeral muscular dystrophy (FSHD) represents a major unmet clinical need arising from the progressive weakness and atrophy of skeletal muscles. The dearth of adequate experimental models has severely hampered our understanding of the disease. To date, no treatment is available for FSHD. Human embryonic stem cells (hESCs) potentially represent a renewable source of skeletal muscle cells (SkMCs) and provide an alternative to invasive patient biopsies. We developed a scalable monolayer system to differentiate hESCs into mature SkMCs within 26 days, without cell sorting or genetic manipulation. Here we show that SkMCs derived from FSHD1-affected hESC lines exclusively express the FSHD pathogenic marker double homeobox 4 and exhibit some of the defects reported in FSHD. FSHD1 myotubes are thinner when compared with unaffected and Becker muscular dystrophy myotubes, and differentially regulate genes involved in cell cycle control, oxidative stress response, and cell adhesion. This cellular model will be a powerful tool for studying FSHD and will ultimately assist in the development of effective treatments for muscular dystrophies. SIGNIFICANCE This work describes an efficient and highly scalable monolayer system to differentiate human pluripotent stem cells (hPSCs) into skeletal muscle cells (SkMCs) and demonstrates disease-specific phenotypes in SkMCs derived from both embryonic and induced hPSCs affected with facioscapulohumeral muscular dystrophy. This study represents the first human stem cell-based cellular model for a muscular dystrophy that is suitable for high-throughput screening and drug development.
Collapse
Affiliation(s)
- Leslie Caron
- Genea Biocells Pty. Ltd., Sydney, New South Wales, Australia
| | - Devaki Kher
- Genea Biocells Pty. Ltd., Sydney, New South Wales, Australia
| | - Kian Leong Lee
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Robert McKernan
- Genea Biocells Pty. Ltd., Sydney, New South Wales, Australia
| | | | | | - Jia Li
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Henry Yang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Heather Main
- Genea Biocells Pty. Ltd., Sydney, New South Wales, Australia
| | - Giulia Ferri
- Dulbecco Telethon Institute and Division of Regenerative Medicine, San Raffaele Scientific Institute, Milano, Italy Vita-Salute San Raffaele University, Milano, Italy
| | - Lisa M Petek
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Lorenz Poellinger
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Daniel G Miller
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Davide Gabellini
- Dulbecco Telethon Institute and Division of Regenerative Medicine, San Raffaele Scientific Institute, Milano, Italy
| | - Uli Schmidt
- Genea Biocells Pty. Ltd., Sydney, New South Wales, Australia Genea Biocells US Inc., San Diego, California, USA
| |
Collapse
|
26
|
Bao B, Maruyama R, Yokota T. Targeting mRNA for the treatment of facioscapulohumeral muscular dystrophy. Intractable Rare Dis Res 2016; 5:168-76. [PMID: 27672539 PMCID: PMC4995414 DOI: 10.5582/irdr.2016.01056] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is an inherited autosomal dominant disorder characterized clinically by progressive muscle degeneration. Currently, no curative treatment for this disorder exists. FSHD patients are managed through physiotherapy to improve function and quality of life. Over the last two decades, FSHD has been better understood as a disease genetically characterized by a pathogenic contraction of a subset of macrosatellite repeats on chromosome 4. Specifically, several studies support an FSHD pathogenesis model involving the aberrant expression of the double homeobox protein 4 (DUX4) gene. Hence, potential therapies revolving around inhibition of DUX4 have been explored. One of the potential treatment options is the use of effective antisense oligonucleotides (AOs) to knockdown expression of the myopathic DUX4 gene and its downstream molecules including paired-like homeodomain transcription factor 1 (PITX1). Success in the suppression of PITX1 expression has already been demonstrated systemically in vivo in recent studies. In this article, we will review the pathogenesis of FSHD and the latest research involving the use of antisense knockdown therapy.
Collapse
Affiliation(s)
- Bo Bao
- Department of Medical Genetics, School of Human Development, Faculty of Medicine and Dentistry, University of Alberta, Edmonton AB, Canada
| | - Rika Maruyama
- Department of Medical Genetics, School of Human Development, Faculty of Medicine and Dentistry, University of Alberta, Edmonton AB, Canada
| | - Toshifumi Yokota
- Department of Medical Genetics, School of Human Development, Faculty of Medicine and Dentistry, University of Alberta, Edmonton AB, Canada
- Muscular Dystrophy Canada Research Chair, University of Alberta, Edmonton AB, Canada
- Address correspondence to: Dr. Toshifumi Yokota; Department of Medical Genetics, School of Human Development, Faculty of Medicine and Dentistry, University of Alberta, Edmonton AB, Canada T6G 2H7. E-mail:
| |
Collapse
|
27
|
|
28
|
Zhuang P, Zhang J, Wang Y, Zhang M, Song L, Lu Z, Zhang L, Zhang F, Wang J, Zhang Y, Wei H, Li H. Reversal of muscle atrophy by Zhimu and Huangbai herb pair via activation of IGF-1/Akt and autophagy signal in cancer cachexia. Support Care Cancer 2015; 24:1189-98. [DOI: 10.1007/s00520-015-2892-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 08/09/2015] [Indexed: 12/23/2022]
|
29
|
Pandey SN, Khawaja H, Chen YW. Culture Conditions Affect Expression of DUX4 in FSHD Myoblasts. Molecules 2015; 20:8304-15. [PMID: 26007167 PMCID: PMC6272558 DOI: 10.3390/molecules20058304] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 04/28/2015] [Accepted: 04/29/2015] [Indexed: 02/02/2023] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is believed to be caused by aberrant expression of double homeobox 4 (DUX4) due to epigenetic changes of the D4Z4 region at chromosome 4q35. Detecting DUX4 is challenging due to its stochastic expression pattern and low transcription level. In this study, we examined different cDNA synthesis strategies and the sensitivity for DUX4 detection. In addition, we investigated the effects of dexamethasone and knockout serum replacement (KOSR) on DUX4 expression in culture. Our data showed that DUX4 was consistently detected in cDNA samples synthesized using Superscript III. The sensitivity of DUX4 detection was higher in the samples synthesized using oligo(dT) primers compared to random hexamers. Adding dexamethasone to the culture media significantly suppressed DUX4 expression in immortalized (1.3 fold, p < 0.01) and primary (4.7 fold, p < 0.01) FSHD myoblasts, respectively. Culture medium with KOSR increased DUX4 expression and the response is concentration dependent. The findings suggest that detection strategies and culture conditions should be carefully considered when studying DUX4 in cultured cells.
Collapse
Affiliation(s)
- Sachchida Nand Pandey
- Research Center for Genetic Medicine, Children's National Medical Center, Washington, DC 20010, USA.
| | - Hunain Khawaja
- Research Center for Genetic Medicine, Children's National Medical Center, Washington, DC 20010, USA.
| | - Yi-Wen Chen
- Research Center for Genetic Medicine, Children's National Medical Center, Washington, DC 20010, USA.
- Department of Integrative Systems Biology and Department of Pediatrics, George Washington University, Washington, DC 20037, USA.
| |
Collapse
|
30
|
Erekat NS. Apoptotic Mediators are Upregulated in the Skeletal Muscle of Chronic/Progressive Mouse Model of Parkinson's Disease. Anat Rec (Hoboken) 2015; 298:1472-8. [DOI: 10.1002/ar.23124] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 12/22/2014] [Accepted: 12/22/2014] [Indexed: 01/10/2023]
Affiliation(s)
- Nour S. Erekat
- Department of Anatomy; Faculty of Medicine; Jordan University of Science and Technology (JUST); Irbid Jordan
| |
Collapse
|
31
|
Lek A, Rahimov F, Jones PL, Kunkel LM. Emerging preclinical animal models for FSHD. Trends Mol Med 2015; 21:295-306. [PMID: 25801126 DOI: 10.1016/j.molmed.2015.02.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 02/23/2015] [Accepted: 02/25/2015] [Indexed: 12/18/2022]
Abstract
Facioscapulohumeral dystrophy (FSHD) is a unique and complex genetic disease that is not entirely solved. Recent advances in the field have led to a consensus genetic premise for the disorder, enabling researchers to now pursue the design of preclinical models. In this review we explore all available FSHD models (DUX4-dependent and -independent) for their utility in therapeutic discovery and potential to yield novel disease insights. Owing to the complex nature of FSHD, there is currently no single model that accurately recapitulates the genetic and pathophysiological spectrum of the disorder. Existing models emphasize only specific aspects of the disease, highlighting the need for more collaborative research and novel paradigms to advance the translational research space of FSHD.
Collapse
Affiliation(s)
- Angela Lek
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Children's Hospital, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; The Wellstone Program, Departments of Neurology and Cell and Developmental Biology, University of Massachusetts Medical School (UMMS), Worcester, MA 01655, USA.
| | - Fedik Rahimov
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Children's Hospital, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; The Wellstone Program, Departments of Neurology and Cell and Developmental Biology, University of Massachusetts Medical School (UMMS), Worcester, MA 01655, USA
| | - Peter L Jones
- The Wellstone Program, Departments of Neurology and Cell and Developmental Biology, University of Massachusetts Medical School (UMMS), Worcester, MA 01655, USA; The Eunice Kennedy Shriver National Institute of Child Health and Human Development (NIHCD) Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Louis M Kunkel
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Children's Hospital, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; The Wellstone Program, Departments of Neurology and Cell and Developmental Biology, University of Massachusetts Medical School (UMMS), Worcester, MA 01655, USA
| |
Collapse
|
32
|
Feeney SJ, McGrath MJ, Sriratana A, Gehrig SM, Lynch GS, D’Arcy CE, Price JT, McLean CA, Tupler R, Mitchell CA. FHL1 reduces dystrophy in transgenic mice overexpressing FSHD muscular dystrophy region gene 1 (FRG1). PLoS One 2015; 10:e0117665. [PMID: 25695429 PMCID: PMC4335040 DOI: 10.1371/journal.pone.0117665] [Citation(s) in RCA: 7] [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: 09/01/2014] [Accepted: 12/29/2014] [Indexed: 01/01/2023] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is an autosomal-dominant disease with no effective treatment. The genetic cause of FSHD is complex and the primary pathogenic insult underlying the muscle disease is unknown. Several disease candidate genes have been proposed including DUX4 and FRG1. Expression analysis studies of FSHD report the deregulation of genes which mediate myoblast differentiation and fusion. Transgenic mice overexpressing FRG1 recapitulate the FSHD muscular dystrophy phenotype. Our current study selectively examines how increased expression of FRG1 may contribute to myoblast differentiation defects. We generated stable C2C12 cell lines overexpressing FRG1, which exhibited a myoblast fusion defect upon differentiation. To determine if myoblast fusion defects contribute to the FRG1 mouse dystrophic phenotype, this strain was crossed with skeletal muscle specific FHL1-transgenic mice. We previously reported that FHL1 promotes myoblast fusion in vitro and FHL1-transgenic mice develop skeletal muscle hypertrophy. In the current study, FRG1 mice overexpressing FHL1 showed an improvement in the dystrophic phenotype, including a reduced spinal kyphosis, increased muscle mass and myofiber size, and decreased muscle fibrosis. FHL1 expression in FRG1 mice, did not alter satellite cell number or activation, but enhanced myoblast fusion. Primary myoblasts isolated from FRG1 mice showed a myoblast fusion defect that was rescued by FHL1 expression. Therefore, increased FRG1 expression may contribute to a muscular dystrophy phenotype resembling FSHD by impairing myoblast fusion, a defect that can be rescued by enhanced myoblast fusion via expression of FHL1.
Collapse
Affiliation(s)
- Sandra J. Feeney
- Department of Biochemistry & Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia
| | - Meagan J. McGrath
- Department of Biochemistry & Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia
| | - Absorn Sriratana
- Department of Biochemistry & Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia
| | - Stefan M. Gehrig
- Basic and Clinical Myology Laboratory, Department of Physiology, The University of Melbourne, Victoria, 3010, Australia
| | - Gordon S. Lynch
- Basic and Clinical Myology Laboratory, Department of Physiology, The University of Melbourne, Victoria, 3010, Australia
| | - Colleen E. D’Arcy
- Department of Biochemistry & Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia
| | - John T. Price
- Department of Biochemistry & Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia
- Centre for Chronic Disease Prevention and Management, College of Health and Biomedicine, Victoria University, Melbourne, Victoria, 8001, Australia
| | - Catriona A. McLean
- Department of Anatomical Pathology, Alfred Hospital, Prahran, Victoria, 3004, Australia
- Department of Medicine, Central Clinical School, Monash University, Clayton, VIC, 3800, Australia
| | - Rossella Tupler
- Program in Gene Function and Expression, University of Massachusetts Medical School, Worcester, MA, 01655, United States of America
- Dipartimento di Scienze della Vita, Universita di Modena e Reggio Emilia, 41125, Modena, Italy
| | - Christina A. Mitchell
- Department of Biochemistry & Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia
- * E-mail:
| |
Collapse
|
33
|
Homma S, Beermann ML, Boyce FM, Miller JB. Expression of FSHD-related DUX4-FL alters proteostasis and induces TDP-43 aggregation. Ann Clin Transl Neurol 2015; 2:151-66. [PMID: 25750920 PMCID: PMC4338956 DOI: 10.1002/acn3.158] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 11/17/2014] [Accepted: 11/18/2014] [Indexed: 11/17/2022] Open
Abstract
Objective Pathogenesis in facioscapulohumeral muscular dystrophy (FSHD) appears to be due to aberrant expression, particularly in skeletal muscle nuclei, of the full-length isoform of DUX4 (DUX4-FL). Expression of DUX4-FL is known to alter gene expression and to be cytotoxic, but cell responses to DUX4-FL are not fully understood. Our study was designed to identify cellular mechanisms of pathogenesis caused by DUX4-FL expression. Methods We used human myogenic cell cultures to analyze the effects of DUX4-FL when it was expressed either from its endogenous promoter in FSHD cells or by exogenous expression using BacMam vectors. We focused on determining the effects of DUX4-FL on protein ubiquitination and turnover and on aggregation of TDP-43. Results Human FSHD myotubes with endogenous DUX4-FL expression showed both altered nuclear and cytoplasmic distributions of ubiquitinated proteins and aggregation of TDP-43 in DUX4-FL-expressing nuclei. Similar changes were found upon exogenous expression of DUX4-FL, but were not seen upon expression of the non-toxic short isoform DUX4-S. DUX4-FL expression also inhibited protein turnover in a model system and increased the amounts of insoluble ubiquitinated proteins and insoluble TDP-43. Finally, inhibition of the ubiquitin–proteasome system with MG132 produced TDP-43 aggregation similar to DUX4-FL expression. Interpretations Our results identify DUX4-FL-induced inhibition of protein turnover and aggregation of TDP-43, which are pathological changes also found in diseases such as amyotrophic lateral sclerosis and inclusion body myopathy, as potential pathological mechanisms in FSHD.
Collapse
Affiliation(s)
- Sachiko Homma
- Neuromuscular Biology & Disease Group, Departments of Neurology and Physiology & Biophysics, Boston University School of Medicine Boston, Massachusetts, 02118
| | - Mary Lou Beermann
- Neuromuscular Biology & Disease Group, Departments of Neurology and Physiology & Biophysics, Boston University School of Medicine Boston, Massachusetts, 02118
| | - Frederick M Boyce
- Department of Neurology, Massachusetts General Hospital Boston, Massachusetts, 02114
| | - Jeffrey Boone Miller
- Neuromuscular Biology & Disease Group, Departments of Neurology and Physiology & Biophysics, Boston University School of Medicine Boston, Massachusetts, 02118
| |
Collapse
|
34
|
Nicot R, Hottenstein M, Raoul G, Ferri J, Horton M, Tobias JW, Barton E, Gelé P, Sciote JJ. Nodal pathway genes are down-regulated in facial asymmetry. J Craniofac Surg 2014; 25:e548-55. [PMID: 25364968 PMCID: PMC4224967 DOI: 10.1097/scs.0000000000001076] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
PURPOSE Facial asymmetry is a common comorbid condition in patients with jaw deformation malocclusion. Heritability of malocclusion is advancing rapidly, but very little is known regarding genetic contributions to asymmetry. This study identifies differences in expression of key asymmetry-producing genes that are down-regulated in patients with facial asymmetry. METHODS Masseter muscle samples were collected during bilateral sagittal split osteotomy orthognathic surgery to correct skeletal-based malocclusion. Patients were classified as class II or III and open or deep bite malocclusion with or without facial asymmetry. Muscle samples were analyzed for gene expression differences on Affymetrix HT2.0 microarray global expression chips. RESULTS Overall gene expression was different for asymmetric patients compared with other malocclusion classifications by principal component analysis (P < 0.05). We identified differences in the nodal signaling pathway, which promotes development of mesoderm and endoderm and left-right patterning during embryogenesis. Nodal and Lefty expression was 1.39- to 1.84-fold greater (P < 3.41 × 10), whereas integral membrane Nodal modulators Nomo1,2,3 were -5.63 to -5.81 (P < 3.05 × 10) less in asymmetry subjects. Fold differences among intracellular pathway members were negative in the range of -7.02 to -2.47 (P < 0.003). Finally Pitx2, an upstream effector of Nodal known to influence the size of type II skeletal muscle fibers was also significantly decreased in facial asymmetry (P < 0.05). CONCLUSIONS When facial asymmetry is part of skeletal malocclusion, there are decreases in nodal signaling pathway genes in masseter muscle. This data suggest that the nodal signaling pathway is down-regulated to help promote development of asymmetry. Pitx2 expression differences also contributed to both skeletal and muscle development in this condition.
Collapse
Affiliation(s)
- Romain Nicot
- From the *Oral and Maxillofacial Surgery, Université Lille Nord de France, UDSL, Controlled Drug Delivery Systems and Biomaterials, Lille France; †Department of Orthodontics, Temple University; and ‡Molecular Profiling Core, Pennsylvania Center for Musculoskeletal Disorders, Perelman School of Medicine, and §Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and ∥Biological Resources Centre, Clinical Investigation Centre, Regional Hospital Center, University of Lille, Lille France
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Reversal of muscle atrophy by Zhimu-Huangbai herb-pair via Akt/mTOR/FoxO3 signal pathway in streptozotocin-induced diabetic mice. PLoS One 2014; 9:e100918. [PMID: 24968071 PMCID: PMC4072704 DOI: 10.1371/journal.pone.0100918] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 06/02/2014] [Indexed: 12/22/2022] Open
Abstract
Skeletal muscle atrophy is one of the serious complications of diabetes. Zhimu-Huangbai herb-pair (ZB) is widely used in Chinese traditional medicine formulas for treating Xiaoke (known as diabetes) and its complications. However, the effect of ZB on reversal of muscle atrophy and the underlying mechanisms remain unknown. In this research, we investigated the effect and possible mechanisms of ZB on skeletal muscle atrophy in diabetic mice. Animal model of diabetic muscle atrophy was developed by high fat diet (HFD) feeding plus streptozotocin (STZ) injection. After oral adminstration of ZB for 6 weeks, the effects of ZB on reversal of muscle atrophy and the underlying mechanisms were evaluated by biochemical, histological and western blot methods. The skeletal muscle weight, strength, and cross-sectional area of diabetic mice were significantly increased by ZB treatment. Biochemical results showed that ZB treatment reduced the serum glucose level, and elevated the serum insulin-like growth factor 1 (IGF-1) and insulin levels significantly compared with untreated diabetic group. The western blot results showed that ZB activated the mTOR signal pathway, shown as increased phosphorylations (p-) of Akt, mTOR, Raptor, S6K1 and reduced Foxo3 expression compared with the model group. ZB could reverse muscle atrophy in diabetic mice. This may be through activation of mTOR signaling pathway that promotes protein synthesis, and inactivation foxo3 protein that inhibits protein degradation. These findings suggested that ZB may be considered as a potential candidate drug in treatment of diabetic muscle atrophy.
Collapse
|
36
|
Dmitriev P, Kairov U, Robert T, Barat A, Lazar V, Carnac G, Laoudj-Chenivesse D, Vassetzky YS. Cancer-related genes in the transcription signature of facioscapulohumeral dystrophy myoblasts and myotubes. J Cell Mol Med 2013; 18:208-17. [PMID: 24341522 PMCID: PMC3930408 DOI: 10.1111/jcmm.12182] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 10/08/2013] [Indexed: 01/23/2023] Open
Abstract
Muscular dystrophy is a condition potentially predisposing for cancer; however, currently, only Myotonic dystrophy patients are known to have a higher risk of cancer. Here, we have searched for a link between facioscapulohumeral dystrophy (FSHD) and cancer by comparing published transcriptome signatures of FSHD and various malignant tumours and have found a significant enrichment of cancer-related genes among the genes differentially expressed in FSHD. The analysis has shown that gene expression profiles of FSHD myoblasts and myotubes resemble that of Ewing's sarcoma more than that of other cancer types tested. This is the first study demonstrating a similarity between FSHD and cancer cell expression profiles, a finding that might indicate the existence of a common step in the pathogenesis of these two diseases.
Collapse
Affiliation(s)
- Petr Dmitriev
- UMR8126, Université Paris-Sud 11, CNRS, Institut de cancérologie Gustave Roussy, Villejuif, France; INSERM U1046, Université Montpellier I, Montpellier, France
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Morpholino treatment improves muscle function and pathology of Pitx1 transgenic mice. Mol Ther 2013; 22:390-396. [PMID: 24232919 DOI: 10.1038/mt.2013.263] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 11/04/2013] [Indexed: 11/08/2022] Open
Abstract
Paired-like homeodomain transcription factor 1 (PITX1) was proposed to be part of the disease mechanisms of facioscapulohumeral muscular dystrophy (FSHD). We generated a tet-repressible muscle-specific Pitx1 transgenic mouse model which develops phenotypes of muscular dystrophy after the PITX1 expression is induced. In this study, we attempted to block the translation of PITX1 protein using morpholinos. Three groups of the transgenic mice received intravenous injections of phosphorodiamidate morpholino oligomers (PMO) (100 mg/kg), octaguanidinium dendrimer-conjugated morpholino (vivo-morpholino) (10 mg/kg), or phosphate-buffered saline (PBS) after the PITX1 expression was induced. Immunoblotting data showed that PITX1 expression in the triceps and quadriceps was significantly reduced 70% and 63% by the vivo-morpholino treatment, respectively. Muscle pathology of the mice treated with the vivo-morpholino was improved by showing 44% fewer angular-shaped atrophic myofibers. Muscle function determined by grip strength was significantly improved by the vivo-morpholino treatment. The study showed that systemic delivery of the vivo-morpholino reduced the PITX1 expression and improved the muscle phenotypes. Aberrant expression of DUX4 from the last unit of the D4Z4 array has been proposed to be the cause of FSHD. The findings of this study suggest that the same principle may be applied to suppress the aberrantly expressed DUX4 in FSHD.
Collapse
|
38
|
Narola J, Pandey SN, Glick A, Chen YW. Conditional expression of TGF-β1 in skeletal muscles causes endomysial fibrosis and myofibers atrophy. PLoS One 2013; 8:e79356. [PMID: 24244485 PMCID: PMC3828351 DOI: 10.1371/journal.pone.0079356] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 09/30/2013] [Indexed: 11/18/2022] Open
Abstract
To study the effects of transforming growth factor beta 1 (TGF-β1) on fibrosis and failure of regeneration of skeletal muscles, we generated a tet-repressible muscle-specific TGF-β1 transgenic mouse in which expression of TGF-β1 is controlled by oral doxycycline. The mice developed muscle weakness and atrophy after TGF-β1 over-expression. We defined the group of mice that showed phenotype within 2 weeks as early onset (EO) and the rest as late onset (LO), which allowed us to further examine phenotypic differences between the groups. While only mice in the EO group showed significant muscle weakness, pathological changes including endomysial fibrosis and smaller myofibers were observed in both groups at two weeks after the TGF-β1 was over-expressed. In addition, the size of the myofibers and collagen accumulation were significantly different between the two groups. The amount of latent and active TGF-β1 in the muscle and circulation were significantly higher in the EO group compared to the LO or control groups. The up-regulation of the latent TGF-β1 indicated that endogenous TGF-β1 was induced by the expression of the TGF-β1 transgene. Our studies showed that the primary effects of TGF-β1 over-expression in skeletal muscles are muscle wasting and endomysial fibrosis. In addition, the severity of the pathology is associated with the total amount of TGF-β1 and the expression of endogenous TGF-β1. The findings suggest that an auto-feedback loop of TGF-β1 may contribute to the severity of phenotypes.
Collapse
Affiliation(s)
- Jigna Narola
- Research Center for Genetic Medicine, Children’s National Medical Center, Washington, DC, United States of America
| | - Sachchida Nand Pandey
- Research Center for Genetic Medicine, Children’s National Medical Center, Washington, DC, United States of America
| | - Adam Glick
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Yi-Wen Chen
- Research Center for Genetic Medicine, Children’s National Medical Center, Washington, DC, United States of America
- Department of Integrative Systems Biology and Department of Pediatrics, George Washington University, Washington, DC, United States of America
- * E-mail:
| |
Collapse
|
39
|
Sugano M, Negishi Y, Endo-Takahashi Y, Hamano N, Usui M, Suzuki R, Maruyama K, Aramaki Y, Yamamoto M. Gene delivery to periodontal tissue using Bubble liposomes and ultrasound. J Periodontal Res 2013; 49:398-404. [PMID: 23889504 DOI: 10.1111/jre.12119] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/06/2013] [Indexed: 11/30/2022]
Abstract
BACKGROUND AND OBJECTIVE Periodontitis is the most common inflammatory disease caused by oral biofilm infection. For efficient periodontal treatment, it is important to enhance the outcome of existing regenerative therapies. The physical action of an ultrasound may be able to deliver a therapeutic gene or drugs into the local area of the periodontium being treated for periodontal regeneration. Previously, we developed "Bubble liposomes" as a useful carrier for gene or drug delivery, and reported that delivery efficiency was increased with high-frequency ultrasound in vitro and in vivo. Hence, the aim of the present study was to examine the possibility of delivering genes into gingival tissues using Bubble liposomes and ultrasound. MATERIAL AND METHODS We attempted to deliver naked plasmid DNA encoding luciferase or enhanced green fluorescent protein (EGFP) into the lower labial gingiva of Wistar rats using Bubble liposomes, with or without ultrasound exposure. Ultrasound parameters were optimized for intensity (0-4.0 W/cm(2) ) and exposure time (0-120 s) to establish the most efficient conditions for exposure. The efficacy and duration of gene expression in the gingiva were investigated using a luciferase assay and fluorescence microscopy. RESULTS The strongest relative luciferase activity was observed when rats were treated under the following ultrasound conditions: 2.0 W/cm(2) intensity and 30 s of exposure time. Relative luciferase activity, 1 d after gene delivery, was significantly higher in gingiva treated using Bubble liposomes and ultrasound than in gingiva of the other treatment groups. Histological analysis also showed that distinct EGFP-expressing cells were observed in transfected gingiva when rats were treated under optimized conditions. CONCLUSION From these results, the combination of Bubble liposomes and ultrasound provides an efficient technique for delivering plasmid DNA into the gingiva. This technique can be applied for the delivery of a variety of therapeutic molecules into target tissue, and may serve as a useful treatment strategy for periodontitis.
Collapse
Affiliation(s)
- M Sugano
- Department of Periodontology, Showa University School of Dentistry, Tokyo, Japan; Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Harafuji N, Schneiderat P, Walter MC, Chen YW. miR-411 is up-regulated in FSHD myoblasts and suppresses myogenic factors. Orphanet J Rare Dis 2013; 8:55. [PMID: 23561550 PMCID: PMC3637251 DOI: 10.1186/1750-1172-8-55] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 03/22/2013] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Facioscapulohumeral muscular dystrophy (FSHD) is an autosomal dominant muscle disorder, which is linked to the contraction of the D4Z4 array at chromosome 4q35. Recent studies suggest that this shortening of the D4Z4 array leads to aberrant expression of double homeobox protein 4 (DUX4) and causes FSHD. In addition, misregulation of microRNAs (miRNAs) has been reported in muscular dystrophies including FSHD. In this study, we identified a miRNA that is differentially expressed in FSHD myoblasts and investigated its function. METHODS To identify misregulated miRNAs and their potential targets in FSHD myoblasts, we performed expression profiling of both miRNA and mRNA using TaqMan Human MicroRNA Arrays and Affymetrix Human Genome U133A plus 2.0 microarrays, respectively. In addition, we over-expressed miR-411 in C₂C₁₂ cells to determine the effect of miR-411 on myogenic markers. RESULTS Using miRNA and mRNA expression profiling, we identified 8 miRNAs and 1,502 transcripts that were differentially expressed in FSHD myoblasts during cell proliferation. One of the 8 differentially expressed miRNAs, miR-411, was validated by quantitative RT-PCR in both primary (2.1 fold, p<0.01) and immortalized (2.7 fold, p<0.01) myoblasts. In situ hybridization showed cytoplasmic localization of miR-411 in FSHD myoblasts. By analyzing both miRNA and mRNA data using Partek Genomics Suite, we identified 4 mRNAs potentially regulated by miR-411 including YY1 associated factor 2 (YAF2). The down-regulation of YAF2 in immortalized myoblasts was validated by immunoblotting (-3.7 fold, p<0.01). C₂C₁₂ cells were transfected with miR-411 to determine whether miR-411 affects YAF2 expression in myoblasts. The results showed that over-expression of miR-411 reduced YAF2 mRNA expression. In addition, expression of myogenic markers including Myod, myogenin, and myosin heavy chain 1 (Myh1) were suppressed by miR-411. CONCLUSIONS The study demonstrated that miR-411 was differentially expressed in FSHD myoblasts and may play a role in regulating myogenesis.
Collapse
Affiliation(s)
- Naoe Harafuji
- Center for Genetic Medicine Research, Children’s Research Institute, Washington, DC, USA
| | - Peter Schneiderat
- Friedrich-Baur-Institute, Department of Neurology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Maggie C Walter
- Friedrich-Baur-Institute, Department of Neurology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Yi-Wen Chen
- Center for Genetic Medicine Research, Children’s Research Institute, Washington, DC, USA
- Department of Integrative Systems Biology and Department of Pediatrics, George Washington University, Washington, DC, USA
- Center for Genetic Medicine Research, Children’s National Medical Center, 111 Michigan Avenue, NW, Washington, DC 20010, USA
| |
Collapse
|
41
|
Knopp P, Figeac N, Fortier M, Moyle L, Zammit PS. Pitx genes are redeployed in adult myogenesis where they can act to promote myogenic differentiation in muscle satellite cells. Dev Biol 2013; 377:293-304. [PMID: 23438814 DOI: 10.1016/j.ydbio.2013.02.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Revised: 01/29/2013] [Accepted: 02/15/2013] [Indexed: 11/17/2022]
Abstract
Skeletal muscle retains a resident stem cell population called satellite cells. Although mitotically quiescent in mature muscle, satellite cells can be activated to produce myoblast progeny to generate myonuclei for skeletal muscle homoeostasis, hypertrophy and repair. Regulation of satellite cell function in adult requires redeployment of many of the regulatory networks fundamental to developmental myogenesis. Involved in such control of muscle stem cell fate in embryos are members of the Pitx gene family of bicoid-class homeodomain proteins. Here, we investigated the expression and function of all three Pitx genes in muscle satellite cells of adult mice. Endogenous Pitx1 was undetectable, whilst Pitx2a, Pitx2b and Pitx2c were at low levels in proliferating satellite cells, but increased during the early stages of myogenic differentiation. By contrast, proliferating satellite cells expressed robust amounts of Pitx3, with levels then decreasing as cells differentiated, although Pitx3 remained expressed in unfused myoblasts. To examine the role of Pitx genes in satellite cell function, retroviral-mediated expression of Pitx1, all Pitx2 isoforms or Pitx3, was used. Constitutive expression of any Pitx isoform suppressed satellite cell proliferation, with the cells undergoing enhanced myogenic differentiation. Conversely, myogenic differentiation into multinucleated myotubes was decreased when Pitx2 or Pitx3 levels were reduced using siRNA. Together, our results show that Pitx genes play a role in regulating satellite cell function during myogenesis in adult.
Collapse
Affiliation(s)
- Paul Knopp
- King's College London, Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, London, SE1 1UL, England, UK
| | | | | | | | | |
Collapse
|