1
|
Sun Z, Wang X, White Z, Dormuth C, Morales F, Bernatchez P. Dyslipidemia in Muscular Dystrophy: A Systematic Review and Meta-Analysis. J Neuromuscul Dis 2023:JND230064. [PMID: 37182897 DOI: 10.3233/jnd-230064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
BACKGROUND Muscular dystrophies (MDs) are characterized by chronic muscle wasting but also poorly understood metabolic co-morbidities. We have recently shown that Duchenne MD (DMD) patients, dogs and asymptomatic carriers are affected by a new form of dyslipidemia that may exacerbate muscle damage. OBJECTIVE We aimed to perform a systematic review and meta-analysis for evidence that other types of MDs are associated with dyslipidemia compared to healthy controls. METHODS Search was conducted using MEDLINE, EMBASE, Cochrane Central Register of Controlled Trials for reports that compare plasma/serum lipids from MD patients and controls, and meta-analysis of cross-sectional studies quantifying total cholesterol, high-density lipoprotein, low density lipoprotein and triglycerides was performed. RESULTS Out of 749 studies, 17 met our inclusion criteria for meta-analysis. 14 of the 17 studies (82% ) included investigated myotonic dystrophy (DM); other studies were on pseudohypertrophic MD (PMD) or DMD. As a whole, MD individuals had significantly higher levels of circulating total cholesterol (Hedges' g with 95% confidence interval [CI], 0.80 [0.03 - 1.56]; p = 0.04) and triglycerides (Hedges' g with 95% confidence interval [CI], 2.28[0.63 - 3.92]; p = 0.01) compared to controls. Meta-regression analysis showed the percentage of male gender was significantly associated with the difference in total cholesterol (beta = 0.05; 95% CI, - 0.02 to 0.11; p = 0.043) and high-density lipoprotein (beta = - 9.38; 95% CI, - 16.26 to - 2.50; p = 0.028). CONCLUSIONS MD is associated with significantly higher circulating levels of total cholesterol and triglycerides. However, caution on the interpretation of these findings is warranted and future longitudinal research is required to better understand this relationship.
Collapse
Affiliation(s)
- Zeren Sun
- University of British Columbia (UBC) Department of Anesthesiology, Pharmacology & Therapeutics, Vancouver, Canada
- UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, Canada
| | - Xindi Wang
- University of British Columbia (UBC) Department of Anesthesiology, Pharmacology & Therapeutics, Vancouver, Canada
- UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, Canada
| | - Zoe White
- University of British Columbia (UBC) Department of Anesthesiology, Pharmacology & Therapeutics, Vancouver, Canada
- UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, Canada
| | - Colin Dormuth
- University of British Columbia (UBC) Department of Anesthesiology, Pharmacology & Therapeutics, Vancouver, Canada
| | - Fernando Morales
- Instituto de Investigaciones en Salud (INISA), Universidad de Costa Rica, SanJosé, Costa Rica
| | - Pascal Bernatchez
- University of British Columbia (UBC) Department of Anesthesiology, Pharmacology & Therapeutics, Vancouver, Canada
- UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, Canada
| |
Collapse
|
2
|
Costa A, Cruz AC, Martins F, Rebelo S. Protein Phosphorylation Alterations in Myotonic Dystrophy Type 1: A Systematic Review. Int J Mol Sci 2023; 24:ijms24043091. [PMID: 36834509 PMCID: PMC9965115 DOI: 10.3390/ijms24043091] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/21/2023] [Accepted: 02/01/2023] [Indexed: 02/09/2023] Open
Abstract
Among the most common muscular dystrophies in adults is Myotonic Dystrophy type 1 (DM1), an autosomal dominant disorder characterized by myotonia, muscle wasting and weakness, and multisystemic dysfunctions. This disorder is caused by an abnormal expansion of the CTG triplet at the DMPK gene that, when transcribed to expanded mRNA, can lead to RNA toxic gain of function, alternative splicing impairments, and dysfunction of different signaling pathways, many regulated by protein phosphorylation. In order to deeply characterize the protein phosphorylation alterations in DM1, a systematic review was conducted through PubMed and Web of Science databases. From a total of 962 articles screened, 41 were included for qualitative analysis, where we retrieved information about total and phosphorylated levels of protein kinases, protein phosphatases, and phosphoproteins in DM1 human samples and animal and cell models. Twenty-nine kinases, 3 phosphatases, and 17 phosphoproteins were reported altered in DM1. Signaling pathways that regulate cell functions such as glucose metabolism, cell cycle, myogenesis, and apoptosis were impaired, as seen by significant alterations to pathways such as AKT/mTOR, MEK/ERK, PKC/CUGBP1, AMPK, and others in DM1 samples. This explains the complexity of DM1 and its different manifestations and symptoms, such as increased insulin resistance and cancer risk. Further studies can be done to complement and explore in detail specific pathways and how their regulation is altered in DM1, to find what key phosphorylation alterations are responsible for these manifestations, and ultimately to find therapeutic targets for future treatments.
Collapse
|
3
|
De Serres-Bérard T, Ait Benichou S, Jauvin D, Boutjdir M, Puymirat J, Chahine M. Recent Progress and Challenges in the Development of Antisense Therapies for Myotonic Dystrophy Type 1. Int J Mol Sci 2022; 23:13359. [PMID: 36362145 PMCID: PMC9657934 DOI: 10.3390/ijms232113359] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/20/2022] [Accepted: 10/25/2022] [Indexed: 08/01/2023] Open
Abstract
Myotonic dystrophy type 1 (DM1) is a dominant genetic disease in which the expansion of long CTG trinucleotides in the 3' UTR of the myotonic dystrophy protein kinase (DMPK) gene results in toxic RNA gain-of-function and gene mis-splicing affecting mainly the muscles, the heart, and the brain. The CUG-expanded transcripts are a suitable target for the development of antisense oligonucleotide (ASO) therapies. Various chemical modifications of the sugar-phosphate backbone have been reported to significantly enhance the affinity of ASOs for RNA and their resistance to nucleases, making it possible to reverse DM1-like symptoms following systemic administration in different transgenic mouse models. However, specific tissue delivery remains to be improved to achieve significant clinical outcomes in humans. Several strategies, including ASO conjugation to cell-penetrating peptides, fatty acids, or monoclonal antibodies, have recently been shown to improve potency in muscle and cardiac tissues in mice. Moreover, intrathecal administration of ASOs may be an advantageous complementary administration route to bypass the blood-brain barrier and correct defects of the central nervous system in DM1. This review describes the evolution of the chemical design of antisense oligonucleotides targeting CUG-expanded mRNAs and how recent advances in the field may be game-changing by forwarding laboratory findings into clinical research and treatments for DM1 and other microsatellite diseases.
Collapse
Affiliation(s)
- Thiéry De Serres-Bérard
- CERVO Research Center, Institut Universitaire en Santé Mentale de Québec, Quebec City, QC G1J 2G3, Canada
| | - Siham Ait Benichou
- LOEX, CHU de Québec-Université Laval Research Center, Quebec City, QC G1J 1Z4, Canada
| | - Dominic Jauvin
- CERVO Research Center, Institut Universitaire en Santé Mentale de Québec, Quebec City, QC G1J 2G3, Canada
| | - Mohamed Boutjdir
- Cardiovascular Research Program, VA New York Harbor Healthcare System, New York, NY 11209, USA
- Department of Medicine, Cell Biology and Pharmacology, State University of New York Downstate Health Science University, New York, NY 11203, USA
- Department of Medicine, NYU School of Medicine, New York, NY 10016, USA
| | - Jack Puymirat
- LOEX, CHU de Québec-Université Laval Research Center, Quebec City, QC G1J 1Z4, Canada
- Department of Medicine, Faculty of Medicine, Université Laval, Quebec City, QC G1V 0A6, Canada
| | - Mohamed Chahine
- CERVO Research Center, Institut Universitaire en Santé Mentale de Québec, Quebec City, QC G1J 2G3, Canada
- Department of Medicine, Faculty of Medicine, Université Laval, Quebec City, QC G1V 0A6, Canada
| |
Collapse
|
4
|
Scutifero M, Lanza M, Petillo R, De Bernardo M, Passamano L, Rosa N, Politano L. Gender effect on onset, prevalence and surgical treatment of cataract in patients with Myotonic Dystrophy type 1. ACTA MYOLOGICA : MYOPATHIES AND CARDIOMYOPATHIES : OFFICIAL JOURNAL OF THE MEDITERRANEAN SOCIETY OF MYOLOGY 2022; 41:105-110. [PMID: 36349183 PMCID: PMC9628803 DOI: 10.36185/2532-1900-n75] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 09/21/2022] [Indexed: 11/21/2022]
Abstract
Myotonic Dystrophy type 1 (DM1) is the most common muscular dystrophy in adults, affecting 1:8000 individuals. It is a multi-systemic disorder involving muscle, heart, endocrine and respiratory apparatus and eye. The eye symptoms can include ptosis, external ophthalmoplegia, epiphora, and early onset cataracts. Cataracts occur at a much earlier age (usually between 30 and 40) than the general population, where females are usually affected more than men. We studied gender differences in cataract prevalence and treatment age in 243 DM1 patients (134 M; 109 F), aged 18 to 70 years, who were subsequently screened at routine follow-up. For each patient, information was collected on age, sex, CTG expansion, age of cataract onset, and age at cataract surgery, when available. Seventy-three patients, 30 females and 43 males, had cataracts, at a mean age of onset of 41.14 ± 12.64 in females, and 40.36 ± 10.03 in males. Sixty-nine of them underwent cataract surgery, males at an earlier age than females (42.8 ± 9.8 years versus 47.3 ± 12.6 years) and in 52.5% of cases before the age of 40, compared to 17.2% of females. The difference was statistically significant. The assumption that females in general and those with DM1 in particular develop cataracts more frequently and earlier than males is not confirmed, at least in this study. A possible explanation for these results could be related to non-advanced age, the protective role of estrogen and the lower prevalence of smoking in the study population.
Collapse
Affiliation(s)
- Marianna Scutifero
- Cardiomyology and Medical Genetics,
University Hospital of Campania “Luigi
Vanvitelli”, Naples, Italy
| | - Michele Lanza
- Eye Department, University of Campania
“Luigi Vanvitelli”, Naples,
Italy
| | - Roberta Petillo
- Cardiomyology and Medical Genetics,
University Hospital of Campania “Luigi
Vanvitelli”, Naples, Italy
| | | | - Luigia Passamano
- Cardiomyology and Medical Genetics,
University Hospital of Campania “Luigi
Vanvitelli”, Naples, Italy
| | - Nicola Rosa
- Department of Medicine and Surgery,
University of Salerno, Salerno,
Italy
| | - Luisa Politano
- Cardiomyology and Medical Genetics,
University Hospital of Campania “Luigi
Vanvitelli”, Naples, Italy,Correspondence Luisa Politano Cardiomyology and
Medical Genetics, University Hospital of Campania “Luigi
Vanvitelli”, piazza Miraglia 2, 80138 Naples, Italy. E-mail:
| |
Collapse
|
5
|
Delay of EGF-Stimulated EGFR Degradation in Myotonic Dystrophy Type 1 (DM1). Cells 2022; 11:cells11193018. [PMID: 36230978 PMCID: PMC9562898 DOI: 10.3390/cells11193018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 09/02/2022] [Accepted: 09/22/2022] [Indexed: 11/18/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1) is an autosomal dominant disease caused by a CTG repeat expansion in the 3′ untranslated region of the dystrophia myotonica protein kinase gene. AKT dephosphorylation and autophagy are associated with DM1. Autophagy has been widely studied in DM1, although the endocytic pathway has not. AKT has a critical role in endocytosis, and its phosphorylation is mediated by the activation of tyrosine kinase receptors, such as epidermal growth factor receptor (EGFR). EGF-activated EGFR triggers the internalization and degradation of ligand–receptor complexes that serve as a PI3K/AKT signaling platform. Here, we used primary fibroblasts from healthy subjects and DM1 patients. DM1-derived fibroblasts showed increased autophagy flux, with enlarged endosomes and lysosomes. Thereafter, cells were stimulated with a high concentration of EGF to promote EGFR internalization and degradation. Interestingly, EGF binding to EGFR was reduced in DM1 cells and EGFR internalization was also slowed during the early steps of endocytosis. However, EGF-activated EGFR enhanced AKT and ERK1/2 phosphorylation levels in the DM1-derived fibroblasts. Therefore, there was a delay in EGF-stimulated EGFR endocytosis in DM1 cells; this alteration might be due to the decrease in the binding of EGF to EGFR, and not to a decrease in AKT phosphorylation.
Collapse
|
6
|
Molecular Therapies for Myotonic Dystrophy Type 1: From Small Drugs to Gene Editing. Int J Mol Sci 2022; 23:ijms23094622. [PMID: 35563013 PMCID: PMC9101876 DOI: 10.3390/ijms23094622] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 12/16/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1) is the most common muscular dystrophy affecting many different body tissues, predominantly skeletal and cardiac muscles and the central nervous system. The expansion of CTG repeats in the DM1 protein-kinase (DMPK) gene is the genetic cause of the disease. The pathogenetic mechanisms are mainly mediated by the production of a toxic expanded CUG transcript from the DMPK gene. With the availability of new knowledge, disease models, and technical tools, much progress has been made in the discovery of altered pathways and in the potential of therapeutic intervention, making the path to the clinic a closer reality. In this review, we describe and discuss the molecular therapeutic strategies for DM1, which are designed to directly target the CTG genomic tract, the expanded CUG transcript or downstream signaling molecules.
Collapse
|
7
|
Thomas MT, Shah S, Popat H, Hanna B, Jani P. Hypoglycaemia and myotonic dystrophy. J Paediatr Child Health 2022; 58:713-714. [PMID: 34184806 DOI: 10.1111/jpc.15633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/09/2021] [Accepted: 06/13/2021] [Indexed: 11/30/2022]
Affiliation(s)
- Meryl T Thomas
- Neonatal Intensive Care Unit, Westmead Hospital, Sydney, New South Wales, Australia
| | - Swapnil Shah
- Neonatal intensive care unit, Westmead Hospital, Sydney, New South Wales, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Himanshu Popat
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Grace Centre for Newborn Care, The Children's Hospital Westmead, Sydney, New South Wales, Australia
| | - Bernadette Hanna
- Clinical Genetics, The Children's Hospital Westmead, Sydney, New South Wales, Australia
| | - Pranav Jani
- Neonatal intensive care unit, Westmead Hospital, Sydney, New South Wales, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
8
|
Pei J, Wang B, Wang D. Current Studies on Molecular Mechanisms of Insulin Resistance. J Diabetes Res 2022; 2022:1863429. [PMID: 36589630 PMCID: PMC9803571 DOI: 10.1155/2022/1863429] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 06/06/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
Diabetes is a metabolic disease that raises the risk of microvascular and neurological disorders. Insensitivity to insulin is a characteristic of type II diabetes, which accounts for 85-90 percent of all diabetic patients. The fundamental molecular factor of insulin resistance may be impaired cell signal transduction mediated by the insulin receptor (IR). Several cell-signaling proteins, including IR, insulin receptor substrate (IRS), and phosphatidylinositol 3-kinase (PI3K), have been recognized as being important in the impaired insulin signaling pathway since they are associated with a large number of proteins that are strictly regulated and interact with other signaling pathways. Many studies have found a correlation between IR alternative splicing, IRS gene polymorphism, the complicated regulatory function of IRS serine/threonine phosphorylation, and the negative regulatory role of p85 in insulin resistance and diabetes mellitus. This review brings up-to-date knowledge of the roles of signaling proteins in insulin resistance in order to aid in the discovery of prospective targets for insulin resistance treatment.
Collapse
Affiliation(s)
- Jinli Pei
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Baochun Wang
- The First Department of Gastrointestinal Surgery, Hainan General Hospital, Haikou, Hainan 570228, China
| | - Dayong Wang
- Laboratory of Biopharmaceuticals and Molecular Pharmacology, School of Pharmaceutical Sciences, Hainan University, Hainan 570228, China
- State Key Laboratory of Tropical Biological Resources of the Ministry of Education of China, Hainan University, Hainan 570228, China
| |
Collapse
|
9
|
Miele L, Perna A, Dajko M, Zocco MA, De Magistris A, Nicoletti TF, Biolato M, Marrone G, Liguori A, Maccora D, Valenza V, Rossi S, Riso V, Di Natale D, Gasbarrini A, Grieco A, Silvestri G. Clinical characteristics of metabolic associated fatty liver disease (MAFLD) in subjects with myotonic dystrophy type 1 (DM1). Dig Liver Dis 2021; 53:1451-1457. [PMID: 33436321 DOI: 10.1016/j.dld.2020.12.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Myotonic dystrophy type 1 (DM1) is a rare inherited neuromuscular disease associated with insulin resistance, and its association with metabolically associated fatty liver disease (MAFLD) has never been explored in prospective studies. The aim of this study was to assess the clinical features of MAFLD in DM1 patients. METHODS We investigated the prevalence and the diagnostic features of MAFLD in a cohort of 29 outpatient fully characterized DM1 patients; afterward, we compared the selected cohort of DM1-MAFLD individuals with a propensity-matched cohort of non-DM1-MAFLD RESULTS: 13/29 (44.83%) DM1 patients received a clinical diagnosis of MAFLD. Compared to DM1 patients with normal liver, DM1-MAFLD individuals showed a higher male prevalence (p = 0.008), BMI (p = 0.014), HOMA score (p = 0.012), and GGT levels (p = 0.050). The statistical comparison showed that the DM1-MAFLD group had a more severe MAFLD according to the FIB4 score than non-DM1-MAFLD patients. This association of a more severe form of liver disease with DM1 remained significant after logistic regression analysis (OR: 6.12, 95% CI 1.44- 26.55).
Collapse
Affiliation(s)
- Luca Miele
- Department of Medical and Surgical Sciences. Fondazione Policlinico Gemelli IRCCS, Università Cattolica del S. Cuore, Roma, Italy.
| | - Alessia Perna
- Department of Neuroscience, Fondazione Policlinico Gemelli IRCCS, Università Cattolica del S. Cuore, Roma, Italy
| | - Marianxhela Dajko
- Department of Medical and Surgical Sciences. Fondazione Policlinico Gemelli IRCCS, Università Cattolica del S. Cuore, Roma, Italy
| | - Maria A Zocco
- Department of Medical and Surgical Sciences. Fondazione Policlinico Gemelli IRCCS, Università Cattolica del S. Cuore, Roma, Italy
| | - Antonio De Magistris
- Department of Medical and Surgical Sciences. Fondazione Policlinico Gemelli IRCCS, Università Cattolica del S. Cuore, Roma, Italy
| | - Tommaso F Nicoletti
- Department of Neuroscience, Fondazione Policlinico Gemelli IRCCS, Università Cattolica del S. Cuore, Roma, Italy
| | - Marco Biolato
- Department of Medical and Surgical Sciences. Fondazione Policlinico Gemelli IRCCS, Università Cattolica del S. Cuore, Roma, Italy
| | - Giuseppe Marrone
- Department of Medical and Surgical Sciences. Fondazione Policlinico Gemelli IRCCS, Università Cattolica del S. Cuore, Roma, Italy
| | - Antonio Liguori
- Department of Medical and Surgical Sciences. Fondazione Policlinico Gemelli IRCCS, Università Cattolica del S. Cuore, Roma, Italy
| | - Daria Maccora
- Department of Radiology, Fondazione Policlinico Gemelli IRCCS, Università Cattolica del S. Cuore, Roma, Italy
| | - Venanzio Valenza
- Department of Radiology, Fondazione Policlinico Gemelli IRCCS, Università Cattolica del S. Cuore, Roma, Italy
| | - Salvatore Rossi
- Department of Neuroscience, Fondazione Policlinico Gemelli IRCCS, Università Cattolica del S. Cuore, Roma, Italy
| | - Vittorio Riso
- Department of Neuroscience, Fondazione Policlinico Gemelli IRCCS, Università Cattolica del S. Cuore, Roma, Italy
| | - Daniele Di Natale
- Department of Neuroscience, Fondazione Policlinico Gemelli IRCCS, Università Cattolica del S. Cuore, Roma, Italy
| | - Antonio Gasbarrini
- Department of Medical and Surgical Sciences. Fondazione Policlinico Gemelli IRCCS, Università Cattolica del S. Cuore, Roma, Italy
| | - Antonio Grieco
- Department of Medical and Surgical Sciences. Fondazione Policlinico Gemelli IRCCS, Università Cattolica del S. Cuore, Roma, Italy
| | - Gabriella Silvestri
- Department of Neuroscience, Fondazione Policlinico Gemelli IRCCS, Università Cattolica del S. Cuore, Roma, Italy
| |
Collapse
|
10
|
Fralish Z, Lotz EM, Chavez T, Khodabukus A, Bursac N. Neuromuscular Development and Disease: Learning From in vitro and in vivo Models. Front Cell Dev Biol 2021; 9:764732. [PMID: 34778273 PMCID: PMC8579029 DOI: 10.3389/fcell.2021.764732] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/06/2021] [Indexed: 01/02/2023] Open
Abstract
The neuromuscular junction (NMJ) is a specialized cholinergic synaptic interface between a motor neuron and a skeletal muscle fiber that translates presynaptic electrical impulses into motor function. NMJ formation and maintenance require tightly regulated signaling and cellular communication among motor neurons, myogenic cells, and Schwann cells. Neuromuscular diseases (NMDs) can result in loss of NMJ function and motor input leading to paralysis or even death. Although small animal models have been instrumental in advancing our understanding of the NMJ structure and function, the complexities of studying this multi-tissue system in vivo and poor clinical outcomes of candidate therapies developed in small animal models has driven the need for in vitro models of functional human NMJ to complement animal studies. In this review, we discuss prevailing models of NMDs and highlight the current progress and ongoing challenges in developing human iPSC-derived (hiPSC) 3D cell culture models of functional NMJs. We first review in vivo development of motor neurons, skeletal muscle, Schwann cells, and the NMJ alongside current methods for directing the differentiation of relevant cell types from hiPSCs. We further compare the efficacy of modeling NMDs in animals and human cell culture systems in the context of five NMDs: amyotrophic lateral sclerosis, myasthenia gravis, Duchenne muscular dystrophy, myotonic dystrophy, and Pompe disease. Finally, we discuss further work necessary for hiPSC-derived NMJ models to function as effective personalized NMD platforms.
Collapse
Affiliation(s)
| | | | | | | | - Nenad Bursac
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, United States
| |
Collapse
|
11
|
Abstract
Myotonic dystrophy is a dominantly inherited multisystem disorder that results from increased CTG repeats in the 3' region of the myotonic dystrophy protein kinase gene (DMPK). The mutant DMPK mRNA remains in the nucleus and sequesters RNA-binding proteins, including regulators of mRNA splicing. Myotonic dystrophy is characterized by a highly variable phenotype that includes muscle weakness and myotonia, and the disorder may affect the function of many endocrine glands. DMPK mRNA is expressed in muscle, testis, liver, pituitary, thyroid, and bone; the mutated form leads to disruption of meiosis and an increase in fetal insulin receptor-A relative to adult insulin receptor-B, resulting in adult primary testicular failure and insulin resistance predisposing to diabetes, respectively. Patients with myotonic dystrophy are also at increased risk for hyperlipidemia, nonalcoholic fatty liver disease, erectile dysfunction, benign and malignant thyroid nodules, bone fractures, miscarriage, preterm delivery, and failed labor during delivery. Circulating parathyroid hormone and adrenocorticotropic hormone levels may be elevated, but the mechanisms for these associations are unclear. This review summarizes what is known about endocrine dysfunction in individuals with myotonic dystrophy.
Collapse
Affiliation(s)
- Stephen J Winters
- Division of Endocrinology, Metabolism and Diabetes, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
12
|
Hintze S, Mensel R, Knaier L, Schoser B, Meinke P. CTG-Repeat Detection in Primary Human Myoblasts of Myotonic Dystrophy Type 1. Front Neurosci 2021; 15:686735. [PMID: 34262431 PMCID: PMC8274452 DOI: 10.3389/fnins.2021.686735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 06/07/2021] [Indexed: 11/18/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1) is an autosomal dominant multisystemic disorder caused by unstable CTG-repeat expansions in the DMPK gene. Tissue mosaicism has been described for the length of these repeat expansions. The most obvious affected tissue is skeletal muscle, making it the first target for therapy development. To date there is no approved therapy despite some existing approaches. Thus, there is the demand to further advance therapeutic developments, which will in return require several well-characterized preclinical tools and model systems. Here we describe a modified method to identify the CTG-repeat length in primary human myoblasts isolated from DM1 patients that requires less genomic DNA and avoids radioactive labeling. Using this method, we show that primary human DM1 myoblast cultures represent a population of cells with different CTG-repeat length. Comparing DNA from the identical muscle biopsy specimen, the range of CTG-repeat length in the myoblast culture is within the same range of the muscle biopsy specimen. In conclusion, primary human DM1 myoblast cultures are a well-suited model to investigate certain aspects of the DM1 pathology. They are a useful platform to perform first-line investigations of preclinical therapies.
Collapse
Affiliation(s)
- Stefan Hintze
- Department of Neurology, LMU Klinikum, Friedrich-Baur-Institute, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Raphaela Mensel
- Department of Neurology, LMU Klinikum, Friedrich-Baur-Institute, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Lisa Knaier
- Department of Neurology, LMU Klinikum, Friedrich-Baur-Institute, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Benedikt Schoser
- Department of Neurology, LMU Klinikum, Friedrich-Baur-Institute, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Peter Meinke
- Department of Neurology, LMU Klinikum, Friedrich-Baur-Institute, Ludwig-Maximilians-University Munich, Munich, Germany
| |
Collapse
|
13
|
Impact of Adenosine Analogue, Adenosine-5'-N-Ethyluronamide (NECA), on Insulin Signaling in Skeletal Muscle Cells. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9979768. [PMID: 34258288 PMCID: PMC8257337 DOI: 10.1155/2021/9979768] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/11/2021] [Accepted: 06/14/2021] [Indexed: 12/29/2022]
Abstract
Materials and Methods Rat L6 skeletal muscle cells were cultured in 25 cm2 flasks. These differentiated cells were treated, and then, quantitative reverse transcription-polymerase chain reaction (qRT-PCR) (probe-based) was used to measure the relative mRNA expression level for metabolic, inflammatory, and nuclear receptor genes including peroxisome proliferator-activated receptor gamma (PGC-1α), carnitine palmitoyl transferase 1 beta (CPT1B), long-chain acyl-CoA de hydrogenase (LCAD), acetyl-CoA carboxylase beta (ACCβ), pyruvate dehydrogenase kinase 4 (PDK4), hexokinase II (HKII), phosphofructokinase (PFK), interleukin-6 (IL-6), and nuclear receptor subfamily 4, group A (NR4A) at different treatment conditions. Results Adenosine-5′-N-ethyluronamide (NECA), a stable adenosine analogue, significantly stimulate inflammatory mediator (IL-6) (p < 0.001) and nuclear receptors (NR4A) (p < 0.05) and significantly modulate metabolic (PFK, LCAD, PGC-1α, and CPT1B) gene expressions in skeletal muscle cells (p < 0.05, p < 0.05, p < 0.001, and p < 0.01, respectively). This present study shows that there is a noteworthy crosstalk between NECA and insulin at various metabolic levels including glycolysis (HKII), fatty acid oxidation (ACCβ), and insulin sensitivity (PDK4). Conclusions A novel crosstalk between adenosine analogue and insulin has been demonstrated for the first time; evidence has been gathered in vitro for the effects of NECA and insulin treatment on intracellular signaling pathways, in particular glycolysis and insulin sensitivity in skeletal muscle cells.
Collapse
|
14
|
Mateus T, Almeida I, Costa A, Viegas D, Magalhães S, Martins F, Herdeiro MT, da Cruz e Silva OAB, Fraga C, Alves I, Nunes A, Rebelo S. Fourier-Transform Infrared Spectroscopy as a Discriminatory Tool for Myotonic Dystrophy Type 1 Metabolism: A Pilot Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18073800. [PMID: 33917301 PMCID: PMC8038712 DOI: 10.3390/ijerph18073800] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/26/2021] [Accepted: 03/30/2021] [Indexed: 12/19/2022]
Abstract
Myotonic dystrophy type 1 (DM1) is a hereditary disease characterized by progressive distal muscle weakness and myotonia. Patients with DM1 have abnormal lipid metabolism and a high propensity to develop a metabolic syndrome in comparison to the general population. It follows that metabolome evaluation in these patients is crucial and may contribute to a better characterization and discrimination between DM1 disease phenotypes and severities. Several experimental approaches are possible to carry out such an analysis; among them is Fourier-transform infrared spectroscopy (FTIR) which evaluates metabolic profiles by categorizing samples through their biochemical composition. In this study, FTIR spectra were acquired and analyzed using multivariate analysis (Principal Component Analysis) using skin DM1 patient-derived fibroblasts and controls. The results obtained showed a clear discrimination between both DM1-derived fibroblasts with different CTG repeat length and with the age of disease onset; this was evident given the distinct metabolic profiles obtained for the two groups. Discrimination could be attributed mainly to the altered lipid metabolism and proteins in the 1800–1500 cm−1 region. These results suggest that FTIR spectroscopy is a valuable tool to discriminate both DM1-derived fibroblasts with different CTG length and age of onset and to study the metabolomic profile of patients with DM1.
Collapse
Affiliation(s)
- Tiago Mateus
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal; (T.M.); (I.A.); (A.C.); (D.V.); (S.M.); (F.M.); (M.T.H.); (O.A.B.d.C.eS.); (A.N.)
| | - Idália Almeida
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal; (T.M.); (I.A.); (A.C.); (D.V.); (S.M.); (F.M.); (M.T.H.); (O.A.B.d.C.eS.); (A.N.)
| | - Adriana Costa
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal; (T.M.); (I.A.); (A.C.); (D.V.); (S.M.); (F.M.); (M.T.H.); (O.A.B.d.C.eS.); (A.N.)
| | - Diana Viegas
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal; (T.M.); (I.A.); (A.C.); (D.V.); (S.M.); (F.M.); (M.T.H.); (O.A.B.d.C.eS.); (A.N.)
| | - Sandra Magalhães
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal; (T.M.); (I.A.); (A.C.); (D.V.); (S.M.); (F.M.); (M.T.H.); (O.A.B.d.C.eS.); (A.N.)
- Department of Chemistry, Aveiro Institute of Materials (CICECO), University of Aveiro, 3810-193 Aveiro, Portugal
| | - Filipa Martins
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal; (T.M.); (I.A.); (A.C.); (D.V.); (S.M.); (F.M.); (M.T.H.); (O.A.B.d.C.eS.); (A.N.)
| | - Maria Teresa Herdeiro
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal; (T.M.); (I.A.); (A.C.); (D.V.); (S.M.); (F.M.); (M.T.H.); (O.A.B.d.C.eS.); (A.N.)
| | - Odete A. B. da Cruz e Silva
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal; (T.M.); (I.A.); (A.C.); (D.V.); (S.M.); (F.M.); (M.T.H.); (O.A.B.d.C.eS.); (A.N.)
| | - Carla Fraga
- Neurology Department, Centro Hospitalar Tâmega e Sousa (CHTS), 4564-007 Penafiel, Portugal; (C.F.); (I.A.)
| | - Ivânia Alves
- Neurology Department, Centro Hospitalar Tâmega e Sousa (CHTS), 4564-007 Penafiel, Portugal; (C.F.); (I.A.)
| | - Alexandra Nunes
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal; (T.M.); (I.A.); (A.C.); (D.V.); (S.M.); (F.M.); (M.T.H.); (O.A.B.d.C.eS.); (A.N.)
| | - Sandra Rebelo
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal; (T.M.); (I.A.); (A.C.); (D.V.); (S.M.); (F.M.); (M.T.H.); (O.A.B.d.C.eS.); (A.N.)
- Correspondence: ; Tel.: +351-924-406-306; Fax: +351-234-372-587
| |
Collapse
|
15
|
Mateus T, Martins F, Nunes A, Herdeiro MT, Rebelo S. Metabolic Alterations in Myotonic Dystrophy Type 1 and Their Correlation with Lipin. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18041794. [PMID: 33673200 PMCID: PMC7918590 DOI: 10.3390/ijerph18041794] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/06/2021] [Accepted: 02/08/2021] [Indexed: 12/14/2022]
Abstract
Myotonic dystrophy type 1 (DM1) is an autosomal dominant hereditary and multisystemic disease, characterized by progressive distal muscle weakness and myotonia. Despite huge efforts, the pathophysiological mechanisms underlying DM1 remain elusive. In this review, the metabolic alterations observed in patients with DM1 and their connection with lipin proteins are discussed. We start by briefly describing the epidemiology, the physiopathological and systemic features of DM1. The molecular mechanisms proposed for DM1 are explored and summarized. An overview of metabolic syndrome, dyslipidemia, and the summary of metabolic alterations observed in patients with DM1 are presented. Patients with DM1 present clinical evidence of metabolic alterations, namely increased levels of triacylglycerol and low-density lipoprotein, increased insulin and glucose levels, increased abdominal obesity, and low levels of high-density lipoprotein. These metabolic alterations may be associated with lipins, which are phosphatidate phosphatase enzymes that regulates the triacylglycerol levels, phospholipids, lipid signaling pathways, and are transcriptional co-activators. Furthermore, lipins are also important for autophagy, inflammasome activation and lipoproteins synthesis. We demonstrate the association of lipin with the metabolic alterations in patients with DM1, which supports further clinical studies and a proper exploration of lipin proteins as therapeutic targets for metabolic syndrome, which is important for controlling many diseases including DM1.
Collapse
Affiliation(s)
| | | | | | | | - Sandra Rebelo
- Correspondence: ; Tel.: +351-924-406-306; Fax: +351-234-372-587
| |
Collapse
|
16
|
Zhang N, Bewick B, Xia G, Furling D, Ashizawa T. A CRISPR-Cas13a Based Strategy That Tracks and Degrades Toxic RNA in Myotonic Dystrophy Type 1. Front Genet 2020; 11:594576. [PMID: 33362853 PMCID: PMC7758406 DOI: 10.3389/fgene.2020.594576] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 11/23/2020] [Indexed: 12/18/2022] Open
Abstract
Cas13a, an effector of type VI CRISPR-Cas systems, is an RNA guided RNase with multiplexing and therapeutic potential. This study employs the Leptotrichia shahii (Lsh) Cas13a and a repeat-based CRISPR RNA (crRNA) to track and eliminate toxic RNA aggregates in myotonic dystrophy type 1 (DM1) – a neuromuscular disease caused by CTG expansion in the DMPK gene. We demonstrate that LshCas13a cleaves CUG repeat RNA in biochemical assays and reduces toxic RNA load in patient-derived myoblasts. As a result, LshCas13a reverses the characteristic adult-to-embryonic missplicing events in several key genes that contribute to DM1 phenotype. The deactivated LshCas13a can further be repurposed to track RNA-rich organelles within cells. Our data highlights the reprogrammability of LshCas13a and the possible use of Cas13a to target expanded repeat sequences in microsatellite expansion diseases.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Neurology, Houston Methodist Research Institute, Houston, TX, United States
| | - Brittani Bewick
- Department of Neurology, Houston Methodist Research Institute, Houston, TX, United States
| | - Guangbin Xia
- Indiana University School of Medicine, Fort Wayne, IN, United States
| | - Denis Furling
- Institut National de la Sante et de la Recherche Medicale (INSERM), Centre de Recherche en Myologie (CRM), Association Institut de Myologie, Sorbonne Université, Paris, France
| | - Tetsuo Ashizawa
- Department of Neurology, Houston Methodist Research Institute, Houston, TX, United States
| |
Collapse
|
17
|
Meola G. Myotonic dystrophy type 2: the 2020 update. ACTA MYOLOGICA : MYOPATHIES AND CARDIOMYOPATHIES : OFFICIAL JOURNAL OF THE MEDITERRANEAN SOCIETY OF MYOLOGY 2020; 39:222-234. [PMID: 33458578 PMCID: PMC7783423 DOI: 10.36185/2532-1900-026] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 11/09/2020] [Indexed: 12/26/2022]
Abstract
The myotonic dystrophies are the commonest cause of adult-onset muscular dystrophy. Phenotypes of DM1 and DM2 are similar, but there are some important differences, including the presence or absence of congenital form, muscles primarily affected (distal vs proximal), involved muscle fiber types (type 1 vs type 2 fibers), and some associated multisystemic phenotypes. There is currently no cure for the myotonic dystrophies but effective management significantly reduces the morbidity and mortality of patients. For the enormous understanding of the molecular pathogenesis of myotonic dystrophy type 1 and myotonic dystrophy type 2, these diseases are now called "spliceopathies" and are mediated by a primary disorder of RNA rather than proteins. Despite clinical and genetic similarities, myotonic dystrophy type 1 and type 2 are distinct disorders requiring different diagnostic and management strategies. Gene therapy for myotonic dystrophy type 1 and myotonic dystrophy type 2 appears to be very close and the near future is an exciting time for clinicians and patients.
Collapse
Affiliation(s)
- Giovanni Meola
- Department of Biomedical Sciences for Health, University of Milan, Italy.,Department of Neurorehabilitation Sciences, Casa di Cura del Policlinico, Milan, Italy
| |
Collapse
|
18
|
Perna A, Maccora D, Rossi S, Nicoletti TF, Zocco MA, Riso V, Modoni A, Petrucci A, Valenza V, Grieco A, Miele L, Silvestri G. High Prevalence and Gender-Related Differences of Gastrointestinal Manifestations in a Cohort of DM1 Patients: A Perspective, Cross-Sectional Study. Front Neurol 2020; 11:394. [PMID: 32595582 PMCID: PMC7303304 DOI: 10.3389/fneur.2020.00394] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 04/17/2020] [Indexed: 12/14/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1, MIM #160900), the most common muscular dystrophy among adults, is a multisystem disorder, which affects, besides the skeletal muscle, several other tissues and/or organs, including the gastrointestinal apparatus, with manifestations that frequently affect the quality of life of DM1 patients. So far, only few, mainly retrospective studies evaluated this specific topic in DM1, so we performed a perspective study, enrolling 61 DM1 patients who underwent an extensive diagnostic protocol, including administration of the Gastrointestinal Symptom Rating Scale (GSRS), a validated patient-reported questionnaire about GI symptoms, laboratory tests, liver US scan, and an intestinal permeability assay, in order to characterize frequency and assess correlations regarding specific gastrointestinal manifestations with demographic or other DM1-related features. Our results in our DM1 cohort confirm the high frequency of various gastrointestinal manifestations, with the most frequent being constipation (45.9%). γGT levels were pathologically increased in 65% of DM1 patients and GPT in 29.82%; liver ultrasound studies showed steatosis in 34.4% of patients. Significantly, 91.22% of DM1 patients showed signs of altered intestinal permeability at the specific assay. We documented a gender-related prevalence and severity of gastrointestinal manifestations in DM1 females compared to DM1 males, while males showed higher serum GPT and γGT levels than females. Correlation studies documented a direct correlation between severity of muscle weakness estimated by MIRS score and γGT and alkaline phosphatase levels, suggesting their potential use as biomarkers of muscle disease severity in DM1.
Collapse
Affiliation(s)
- Alessia Perna
- Institute of Neurology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Daria Maccora
- Department of Image Diagnostics, Oncological Radiotherapy and Hematology Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Salvatore Rossi
- Institute of Neurology, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | - Maria Assunta Zocco
- Department of Internal Medicine, Gastroenterology and Hepatology, Fondazione Policlinico Agostino Gemelli IRCCS, Rome, Italy
| | - Vittorio Riso
- Institute of Neurology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Anna Modoni
- UOC of Neurology, Area of Neuroscience, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Antonio Petrucci
- Center for Neuromuscular and Neurological Rare Disease, S. Camillo Forlanini Hospital, Rome, Italy
| | - Venanzio Valenza
- Department of Image Diagnostics, Oncological Radiotherapy and Hematology Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Antonio Grieco
- Department of Gastroenterological, Endocrine-Metabolic and Nefro-Urological Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Luca Miele
- Department of Gastroenterological, Endocrine-Metabolic and Nefro-Urological Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Gabriella Silvestri
- Institute of Neurology, Università Cattolica del Sacro Cuore, Rome, Italy.,UOC of Neurology, Area of Neuroscience, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
19
|
Nieuwenhuis S, Okkersen K, Widomska J, Blom P, 't Hoen PAC, van Engelen B, Glennon JC. Insulin Signaling as a Key Moderator in Myotonic Dystrophy Type 1. Front Neurol 2019; 10:1229. [PMID: 31849810 PMCID: PMC6901991 DOI: 10.3389/fneur.2019.01229] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 11/05/2019] [Indexed: 12/15/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1) is an autosomal dominant genetic disease characterized by multi-system involvement. Affected organ system includes skeletal muscle, heart, gastro-intestinal system and the brain. In this review, we evaluate the evidence for alterations in insulin signaling and their relation to clinical DM1 features. We start by summarizing the molecular pathophysiology of DM1. Next, an overview of normal insulin signaling physiology is given, and evidence for alterations herein in DM1 is presented. Clinically, evidence for involvement of insulin signaling pathways in DM1 is based on the increased incidence of insulin resistance seen in clinical practice and recent trial evidence of beneficial effects of metformin on muscle function. Indirectly, further support may be derived from certain CNS derived symptoms characteristic of DM1, such as obsessive-compulsive behavior features, for which links with altered insulin signaling has been demonstrated in other diseases. At the basic scientific level, several pathophysiological mechanisms that operate in DM1 may compromise normal insulin signaling physiology. The evidence presented here reflects the importance of insulin signaling in relation to clinical features of DM1 and justifies further basic scientific and clinical, therapeutically oriented research.
Collapse
Affiliation(s)
- Sylvia Nieuwenhuis
- Department of Cognitive Neuroscience, Donders Institute for Brain Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Kees Okkersen
- Department of Neurology, Donders Institute for Brain Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Joanna Widomska
- Department of Cognitive Neuroscience, Donders Institute for Brain Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Paul Blom
- VDL Enabling Technologies Group B.V., Eindhoven, Netherlands
| | - Peter A C 't Hoen
- Centre for Molecular and Biomolecular Informatics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Baziel van Engelen
- Department of Neurology, Donders Institute for Brain Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Jeffrey C Glennon
- Department of Cognitive Neuroscience, Donders Institute for Brain Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, Netherlands
| |
Collapse
|
20
|
López Castel A, Overby SJ, Artero R. MicroRNA-Based Therapeutic Perspectives in Myotonic Dystrophy. Int J Mol Sci 2019; 20:ijms20225600. [PMID: 31717488 PMCID: PMC6888406 DOI: 10.3390/ijms20225600] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/28/2019] [Accepted: 10/30/2019] [Indexed: 12/20/2022] Open
Abstract
Myotonic dystrophy involves two types of chronically debilitating rare neuromuscular diseases: type 1 (DM1) and type 2 (DM2). Both share similarities in molecular cause, clinical signs, and symptoms with DM2 patients usually displaying milder phenotypes. It is well documented that key clinical symptoms in DM are associated with a strong mis-regulation of RNA metabolism observed in patient’s cells. This mis-regulation is triggered by two leading DM-linked events: the sequestration of Muscleblind-like proteins (MBNL) and the mis-regulation of the CUGBP RNA-Binding Protein Elav-Like Family Member 1 (CELF1) that cause significant alterations to their important functions in RNA processing. It has been suggested that DM1 may be treatable through endogenous modulation of the expression of MBNL and CELF1 proteins. In this study, we analyzed the recent identification of the involvement of microRNA (miRNA) molecules in DM and focus on the modulation of these miRNAs to therapeutically restore normal MBNL or CELF1 function. We also discuss additional prospective miRNA targets, the use of miRNAs as disease biomarkers, and additional promising miRNA-based and miRNA-targeting drug development strategies. This review provides a unifying overview of the dispersed data on miRNA available in the context of DM.
Collapse
Affiliation(s)
- Arturo López Castel
- Translational Genomics Group, Incliva Health Research Institute, Burjassot, 46100 Valencia, Spain
- Interdisciplinary Research Structure for Biotechnology and Biomedicine (Eri Biotecmed), University of Valencia, Burjassot, 46100 Valencia, Spain
- Correspondence: (A.L.C.); (R.A.)
| | - Sarah Joann Overby
- Translational Genomics Group, Incliva Health Research Institute, Burjassot, 46100 Valencia, Spain
- Interdisciplinary Research Structure for Biotechnology and Biomedicine (Eri Biotecmed), University of Valencia, Burjassot, 46100 Valencia, Spain
| | - Rubén Artero
- Translational Genomics Group, Incliva Health Research Institute, Burjassot, 46100 Valencia, Spain
- Interdisciplinary Research Structure for Biotechnology and Biomedicine (Eri Biotecmed), University of Valencia, Burjassot, 46100 Valencia, Spain
- Correspondence: (A.L.C.); (R.A.)
| |
Collapse
|
21
|
Sznajder ŁJ, Swanson MS. Short Tandem Repeat Expansions and RNA-Mediated Pathogenesis in Myotonic Dystrophy. Int J Mol Sci 2019; 20:ijms20133365. [PMID: 31323950 PMCID: PMC6651174 DOI: 10.3390/ijms20133365] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 06/27/2019] [Accepted: 07/08/2019] [Indexed: 12/23/2022] Open
Abstract
Short tandem repeat (STR) or microsatellite, expansions underlie more than 50 hereditary neurological, neuromuscular and other diseases, including myotonic dystrophy types 1 (DM1) and 2 (DM2). Current disease models for DM1 and DM2 propose a common pathomechanism, whereby the transcription of mutant DMPK (DM1) and CNBP (DM2) genes results in the synthesis of CUG and CCUG repeat expansion (CUGexp, CCUGexp) RNAs, respectively. These CUGexp and CCUGexp RNAs are toxic since they promote the assembly of ribonucleoprotein (RNP) complexes or RNA foci, leading to sequestration of Muscleblind-like (MBNL) proteins in the nucleus and global dysregulation of the processing, localization and stability of MBNL target RNAs. STR expansion RNAs also form phase-separated gel-like droplets both in vitro and in transiently transfected cells, implicating RNA-RNA multivalent interactions as drivers of RNA foci formation. Importantly, the nucleation and growth of these nuclear foci and transcript misprocessing are reversible processes and thus amenable to therapeutic intervention. In this review, we provide an overview of potential DM1 and DM2 pathomechanisms, followed by a discussion of MBNL functions in RNA processing and how multivalent interactions between expanded STR RNAs and RNA-binding proteins (RBPs) promote RNA foci assembly.
Collapse
Affiliation(s)
- Łukasz J Sznajder
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, FL 32610, USA.
| | - Maurice S Swanson
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, FL 32610, USA
| |
Collapse
|
22
|
Voellenkle C, Perfetti A, Carrara M, Fuschi P, Renna LV, Longo M, Sain SB, Cardani R, Valaperta R, Silvestri G, Legnini I, Bozzoni I, Furling D, Gaetano C, Falcone G, Meola G, Martelli F. Dysregulation of Circular RNAs in Myotonic Dystrophy Type 1. Int J Mol Sci 2019; 20:ijms20081938. [PMID: 31010208 PMCID: PMC6515344 DOI: 10.3390/ijms20081938] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 04/10/2019] [Accepted: 04/17/2019] [Indexed: 01/03/2023] Open
Abstract
Circular RNAs (circRNAs) constitute a recently re-discovered class of non-coding RNAs functioning as sponges for miRNAs and proteins, affecting RNA splicing and regulating transcription. CircRNAs are generated by “back-splicing”, which is the linking covalently of 3′- and 5′-ends of exons. Thus, circRNA levels might be deregulated in conditions associated with altered RNA-splicing. Significantly, growing evidence indicates their role in human diseases. Specifically, myotonic dystrophy type 1 (DM1) is a multisystemic disorder caused by expanded CTG repeats in the DMPK gene which results in abnormal mRNA-splicing. In this investigation, circRNAs expressed in DM1 skeletal muscles were identified by analyzing RNA-sequencing data-sets followed by qPCR validation. In muscle biopsies, out of nine tested, four transcripts showed an increased circular fraction: CDYL, HIPK3, RTN4_03, and ZNF609. Their circular fraction values correlated with skeletal muscle strength and with splicing biomarkers of disease severity, and displayed higher values in more severely affected patients. Moreover, Receiver-Operating-Characteristics curves of these four circRNAs discriminated DM1 patients from controls. The identified circRNAs were also detectable in peripheral-blood-mononuclear-cells (PBMCs) and the plasma of DM1 patients, but they were not regulated significantly. Finally, increased circular fractions of RTN4_03 and ZNF609 were also observed in differentiated myogenic cell lines derived from DM1 patients. In conclusion, this pilot study identified circRNA dysregulation in DM1 patients.
Collapse
Affiliation(s)
- Christine Voellenkle
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy.
| | - Alessandra Perfetti
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy.
| | - Matteo Carrara
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy.
| | - Paola Fuschi
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy.
| | - Laura Valentina Renna
- Laboratory of Muscle Histopathology and Molecular Biology, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy.
| | - Marialucia Longo
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy.
| | - Simona Baghai Sain
- Center for Translational Genomics and Bioinformatics, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.
| | - Rosanna Cardani
- Laboratory of Muscle Histopathology and Molecular Biology, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy.
| | - Rea Valaperta
- Research Laboratories, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy.
| | - Gabriella Silvestri
- Department of Geriatrics, Orthopaedic and Neuroscience, Institute of Neurology, Catholic University of Sacred Heart, Fondazione Policlinico Gemelli, 00168 Rome, Italy.
| | - Ivano Legnini
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy.
| | - Irene Bozzoni
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy.
| | - Denis Furling
- Sorbonne Université, INSERM, Association Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France.
| | - Carlo Gaetano
- Laboratory of Epigenetics, Istituti Clinici Scientifici Maugeri, 27100 Pavia, Italy.
| | - Germana Falcone
- Institute of Cell Biology and Neurobiology, National Research Council, Monterotondo, 00015 Rome, Italy.
| | - Giovanni Meola
- Laboratory of Muscle Histopathology and Molecular Biology, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy.
- Department of Neurology, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy.
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy.
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy.
| |
Collapse
|
23
|
Gonçalves DA, Silveira WA, Manfredi LH, Graça FA, Armani A, Bertaggia E, O Neill BT, Lautherbach N, Machado J, Nogara L, Pereira MG, Arcidiacono D, Realdon S, Kahn CR, Sandri M, Kettelhut IC, Navegantes LCC. Insulin/IGF1 signalling mediates the effects of β 2 -adrenergic agonist on muscle proteostasis and growth. J Cachexia Sarcopenia Muscle 2019; 10:455-475. [PMID: 30932373 PMCID: PMC6463755 DOI: 10.1002/jcsm.12395] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 12/18/2018] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Stimulation of β2 -adrenoceptors can promote muscle hypertrophy and fibre type shift, and it can counteract atrophy and weakness. The underlying mechanisms remain elusive. METHODS Fed wild type (WT), 2-day fasted WT, muscle-specific insulin (INS) receptor (IR) knockout (M-IR-/- ), and MKR mice were studied with regard to acute effects of the β2 -agonist formoterol (FOR) on protein metabolism and signalling events. MKR mice express a dominant negative IGF1 receptor, which blocks both INS/IGF1 signalling. All received one injection of FOR (300 μg kg-1 subcutaneously) or saline. Skeletal muscles and serum samples were analysed from 30 to 240 min. For the study of chronic effects of FOR on muscle plasticity and function as well as intracellular signalling pathways, fed WT and MKR mice were treated with formoterol (300 μg kg-1 day-1 ) for 30 days. RESULTS In fed and fasted mice, one injection of FOR inhibited autophagosome formation (LC3-II content, 65%, P ≤ 0.05) that was paralleled by an increase in serum INS levels (4-fold to 25-fold, P ≤ 0.05) and the phosphorylation of Akt (4.4-fold to 6.5-fold, P ≤ 0.05) and ERK1/2 (50% to two-fold, P ≤ 0.05). This led to the suppression (40-70%, P ≤ 0.05) of the master regulators of atrophy, FoxOs, and the mRNA levels of their target genes. FOR enhanced (41%, P ≤ 0.05) protein synthesis only in fed condition and stimulated (4.4-fold to 35-fold, P ≤ 0.05) the prosynthetic Akt/mTOR/p70S6K pathway in both fed and fasted states. FOR effects on Akt signalling during fasting were blunted in both M-IR-/- and MKR mice. Inhibition of proteolysis markers by FOR was prevented only in MKR mice. Blockade of PI3K/Akt axis and mTORC1, but not ERK1/2, in fasted mice also suppressed the acute FOR effects on proteolysis and autophagy. Chronic stimulation of β2 -adrenoceptors in fed WT mice increased body (11%, P ≤ 0.05) and muscle (15%, P ≤ 0.05) growth and downregulated atrophy-related genes (30-40%, P ≤ 0.05), but these effects were abolished in MKR mice. Increases in muscle force caused by FOR (WT, 24%, P ≤ 0.05) were only partially impaired in MKR mice (12%, P ≤ 0.05), and FOR-induced slow-to-fast fibre type shift was not blocked at all in these animals. In MKR mice, FOR also restored the lower levels of muscle SDH activity to basal WT values and caused a marked reduction (57%, P ≤ 0.05) in the number of centrally nucleated fibers. CONCLUSIONS NS/IGF1 signalling is necessary for the anti-proteolytic and hypertrophic effects of in vivo β2 -adrenergic stimulation and appears to mediate FOR-induced enhancement of protein synthesis. INS/IGF1 signalling only partially contributes to gain in strength and does not mediate fibre type transition induced by FOR.
Collapse
Affiliation(s)
- Dawit A Gonçalves
- Department of Physiology, Ribeirão Preto Medical School/University of São Paulo, Ribeirão Preto, SP, Brazil.,Department of Biochemistry/Immunology, Ribeirão Preto Medical School/University of São Paulo, Ribeirão Preto, SP, Brazil.,Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Wilian A Silveira
- Department of Physiology, Ribeirão Preto Medical School/University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Leandro H Manfredi
- Department of Physiology, Ribeirão Preto Medical School/University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Flávia A Graça
- Department of Physiology, Ribeirão Preto Medical School/University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Andrea Armani
- Department of Biomedical Sciences, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Enrico Bertaggia
- Department of Biomedical Sciences, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Brian T O Neill
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Natalia Lautherbach
- Department of Physiology, Ribeirão Preto Medical School/University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Juliano Machado
- Department of Physiology, Ribeirão Preto Medical School/University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Leonardo Nogara
- Department of Biomedical Sciences, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Marcelo G Pereira
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Diletta Arcidiacono
- Digestive Endoscopy Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Stefano Realdon
- Digestive Endoscopy Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - C Ronald Kahn
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Marco Sandri
- Department of Biomedical Sciences, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy.,Myology Center, University of Padova, Padova, Italy
| | - Isis C Kettelhut
- Department of Physiology, Ribeirão Preto Medical School/University of São Paulo, Ribeirão Preto, SP, Brazil.,Department of Biochemistry/Immunology, Ribeirão Preto Medical School/University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Luiz Carlos C Navegantes
- Department of Physiology, Ribeirão Preto Medical School/University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
24
|
Renna LV, Bosè F, Brigonzi E, Fossati B, Meola G, Cardani R. Aberrant insulin receptor expression is associated with insulin resistance and skeletal muscle atrophy in myotonic dystrophies. PLoS One 2019; 14:e0214254. [PMID: 30901379 PMCID: PMC6430513 DOI: 10.1371/journal.pone.0214254] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 03/08/2019] [Indexed: 12/26/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1) and type 2 (DM2) are autosomal dominant multisystemic disorders linked to two different genetic loci and characterized by several features including myotonia, muscle atrophy and insulin resistance. The aberrant alternative splicing of insulin receptor (IR) gene and post-receptor signalling abnormalities have been associated with insulin resistance, however the precise molecular defects that cause metabolic dysfunctions are still unknown. Thus, the aims of this study were to investigate in DM skeletal muscle biopsies if beyond INSR missplicing, altered IR protein expression could play a role in insulin resistance and to verify if the lack of insulin pathway activation could contribute to skeletal muscle wasting. Our analysis showed that DM skeletal muscle exhibits a lower expression of the insulin receptor in type 1 fibers which can contribute to the defective activation of the insulin pathway. Moreover, the aberrant insulin signalling activation leads to a lower activation of mTOR and to an increase in MuRF1 and Atrogin-1/MAFbx expression, possible explaining DM skeletal muscle fiber atrophy. Taken together our data indicate that the defective insulin signalling activation can contribute to skeletal muscle features in DM patients and are probably linked to an aberrant specific-fiber type expression of the insulin receptor.
Collapse
Affiliation(s)
- Laura Valentina Renna
- Laboratory of Muscle Histopathology and Molecular Biology, IRCCS-Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - Francesca Bosè
- Laboratory of Muscle Histopathology and Molecular Biology, IRCCS-Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - Elisa Brigonzi
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Barbara Fossati
- Department of Neurology, IRCCS-Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - Giovanni Meola
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
- Department of Neurology, IRCCS-Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - Rosanna Cardani
- Laboratory of Muscle Histopathology and Molecular Biology, IRCCS-Policlinico San Donato, San Donato Milanese, Milan, Italy
- * E-mail:
| |
Collapse
|
25
|
Body composition analysis in patients with myotonic dystrophy types 1 and 2. Neurol Sci 2019; 40:1035-1040. [PMID: 30790082 DOI: 10.1007/s10072-019-03763-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 02/13/2019] [Indexed: 10/27/2022]
Abstract
INTRODUCTION To date, there are only several reports on body composition in myotonic dystrophy type 1 (DM1) and there are no data for myotonic dystrophy type 2 (DM2). The aim was to analyze body composition of patients with DM1 and DM2, and its association with socio-demographic and clinical features of the diseases. METHODS There were no statistical differences in sociodemographic features between 20 DM1 patients and 12 DM2 patients. Body composition was assessed by DEXA (dual-energy x-ray absorptiometry). A three-compartment model was used: bone mineral content (BMC), fat mass (FM), and lean tissue mass (LTM). RESULTS Patients with DM1 and DM2 had similar total body mass (TBM), BMC, FM, and LTM. Patients with DM1 had higher trunk-limb fat index (TLFI) in comparison to DM2 patients which indicates visceral fat deposition in DM1 (1.16 ± 0.32 for DM1 vs. 0.87 ± 0.23 for DM2, p < 0.05). Right ribs bone mineral density was lower in DM2 group (0.68 ± 0.07 g/cm2 vs. 0.61 ± 0.09 g/cm2, p < 0.05). Higher percentage of FM in legs showed correlation with lower strength of the upper leg muscles in DM1 (ρ = - 0.47, p < 0.05). Higher muscle strength in DM2 patients was in correlation with higher bone mineral density (ρ = + 0.62, p < 0.05 for upper arm muscles, ρ = + 0.87, p < 0.01 for lower arm muscles, ρ = + 0.72, p < 0.05 for lower leg muscles). CONCLUSION DM1 patients had visceral obesity, and percentage of FM correlated with a degree of muscle weakness in upper legs. In DM2 patients, degree of muscle weakness was in correlation with higher FM index and lower bone mineral density.
Collapse
|
26
|
Souidi A, Zmojdzian M, Jagla K. Dissecting Pathogenetic Mechanisms and Therapeutic Strategies in Drosophila Models of Myotonic Dystrophy Type 1. Int J Mol Sci 2018; 19:E4104. [PMID: 30567354 PMCID: PMC6321436 DOI: 10.3390/ijms19124104] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 12/08/2018] [Accepted: 12/13/2018] [Indexed: 12/16/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1), the most common cause of adult-onset muscular dystrophy, is autosomal dominant, multisystemic disease with characteristic symptoms including myotonia, heart defects, cataracts and testicular atrophy. DM1 disease is being successfully modelled in Drosophila allowing to identify and validate new pathogenic mechanisms and potential therapeutic strategies. Here we provide an overview of insights gained from fruit fly DM1 models, either: (i) fundamental with particular focus on newly identified gene deregulations and their link with DM1 symptoms; or (ii) applied via genetic modifiers and drug screens to identify promising therapeutic targets.
Collapse
Affiliation(s)
- Anissa Souidi
- GReD, INSERM U1103, CNRS, UMR6293, University of Clermont Auvergne, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France.
| | - Monika Zmojdzian
- GReD, INSERM U1103, CNRS, UMR6293, University of Clermont Auvergne, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France.
| | - Krzysztof Jagla
- GReD, INSERM U1103, CNRS, UMR6293, University of Clermont Auvergne, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France.
| |
Collapse
|
27
|
Zhang S, Wei M, Yue M, Wang P, Yin X, Wang L, Yang X, Liu H. Hyperinsulinemia precedes insulin resistance in offspring rats exposed to angiotensin II type 1 autoantibody in utero. Endocrine 2018; 62:588-601. [PMID: 30101377 DOI: 10.1007/s12020-018-1700-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 07/26/2018] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Insulin resistance is highly associated with an adverse intrauterine environment. We previously reported that fetal rats exposed to angiotensin II type 1 receptor (AT1R) autoantibody (AT1-AA) displayed increased susceptibility to metabolic diseases during middle age. However, the timing of the onset of insulin resistance remains unknown. In this study, we examined the offspring of AT1-AA-positive rats, tracking the development of insulin resistance. METHODS Pregnant rats were intravenously injected with AT1-AA. Afterwards, we collected serum samples and liver tissues of the offspring at various stages, including gestation day 18, 3 weeks (weaning period), 18 weeks (young adulthood), and 48 weeks (middle age) after birth. RESULTS Compared with saline control group, hepatic vacuolar degeneration was visible in AT1-AA offspring rats as early as 3 weeks; hyperinsulinemia and impaired glucose tolerance occurred at 18 weeks of age, however, insulin resistance was not observed until 48 weeks. At 18 weeks we detected suppressed protein levels of insulin receptor (IR) but increased levels of IR substrate 1 (IRS1) in the liver of AT1-AA group rats. Interestingly, both IR and IRS1/2 were significantly decreased at 48 weeks. Liver proteomic analysis indicated that the differences in protein expression between the AT1-AA and control rats became more pronounced with age, particularly in terms of mitochondrial energy metabolism. CONCLUSION Rats exposed to AT1-AA in utero developed hyperinsulinemia from young adulthood which subsequently progressed to insulin resistance, and was linked with abnormal hepatic structure and impaired IR signaling. Additionally, dysregulation of energy metabolism may play a fundamental role in predisposing offspring to insulin resistance.
Collapse
Affiliation(s)
- Suli Zhang
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Mingming Wei
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Mingming Yue
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Pengli Wang
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiaochen Yin
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Li Wang
- Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaoli Yang
- Department of Reproductive Center, Taiyuan Central Hospital, Taiyuan, Shanxi, China
| | - Huirong Liu
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Metabolic Disorder Related Cardiovascular Disease, Capital Medical University, Beijing, China.
| |
Collapse
|
28
|
Metabolic impairments in patients with myotonic dystrophy type 2. ACTA MYOLOGICA : MYOPATHIES AND CARDIOMYOPATHIES : OFFICIAL JOURNAL OF THE MEDITERRANEAN SOCIETY OF MYOLOGY 2018; 37:252-256. [PMID: 30944903 PMCID: PMC6416699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVES metabolic syndrome (MetS) increases risk of cardiovascular diseases and diabetes mellitus type 2. Aim of this study was to investigate frequency and features of MetS in a large cohort of patients with DM2. MATERIALS & METHODS this cross-sectional study included 47 DM2 patients. Patients were matched with 94 healthy controls (HCs) for gender and age. MetS was diagnosed according to the new worldwide consensus criteria from 2009. RESULTS mean age of DM2 patients was 52 ± 11 years, 15 (32%) were males, and mean disease duration was 15 ± 14 years. MetS was present in 53% of DM2 patients and 46% of HCs (p > 0.05). All components of the MetS appeared with the similar frequency in DM2 and HCs, respectively: hypertension 64 vs 52%, central obesity 62 vs 74%, hypertriglyceridemia 49 vs 39%, hyperglycemia 42 vs 33% and low HDL cholesterol 30 vs 42% (p > 0.05). DM2 patients were more commonly on lipid lowering therapy compared to HCs (12 vs 3%, p = 0.05). Fifteen (32%) patients with DM2 and only one (1%) subject from control group had diabetes mellitus (p < 0.01). Insulin resistance was found in thirty (65%) patients with DM2. Presence of MetS was not associated with patient's gender, age, severity nor duration of the disease (p > 0.05). CONCLUSIONS more than half of DM2 subjects met the criteria for the MetS. We suppose that treatment of metabolic disturbances may reduce cardiovascular complications and improve quality of life in patients with DM2, which is progressive and still incurable disorder.
Collapse
|
29
|
Matloka M, Klein AF, Rau F, Furling D. Cells of Matter- In Vitro Models for Myotonic Dystrophy. Front Neurol 2018; 9:361. [PMID: 29875732 PMCID: PMC5974047 DOI: 10.3389/fneur.2018.00361] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 05/03/2018] [Indexed: 12/17/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1 also known as Steinert disease) is a multisystemic disorder mainly characterized by myotonia, progressive muscle weakness and wasting, cognitive impairments, and cardiac defects. This autosomal dominant disease is caused by the expression of nuclear retained RNAs containing pathologic expanded CUG repeats that alter the function of RNA-binding proteins in a tissue-specific manner, leading ultimately to neuromuscular dysfunction and clinical symptoms. Although considerable knowledge has been gathered on myotonic dystrophy since its first description, the development of novel relevant disease models remains of high importance to investigate pathophysiologic mechanisms and to assess new therapeutic approaches. In addition to animal models, in vitro cell cultures provide a unique resource for both fundamental and translational research. This review discusses how cellular models broke ground to decipher molecular basis of DM1 and describes currently available cell models, ranging from exogenous expression of the CTG tracts to variable patients' derived cells.
Collapse
Affiliation(s)
| | | | | | - Denis Furling
- Sorbonne Université, INSERM, Association Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| |
Collapse
|