1
|
Bayazit MB, Henderson D, Nguyen KT, Reátegui E, Tawil R, Flanigan KM, Harper SQ, Saad NY. Identification of disease-specific extracellular vesicle-associated plasma protein biomarkers for Duchenne Muscular Dystrophy and Facioscapulohumeral Muscular Dystrophy. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.29.24317861. [PMID: 39649602 PMCID: PMC11623727 DOI: 10.1101/2024.11.29.24317861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Objective Reliable, circulating biomarkers for Duchenne, Becker and facioscapulohumeral muscular dystrophies (DBMD and FSHD) remain unvalidated. Here, we investigated the plasma extracellular vesicle (EV) proteome to identify disease-specific biomarkers that could accelerate therapy approvals. Methods We extracted EVs from the plasma of DBMD and FSHD patients and healthy controls using size-exclusion chromatography, conducted mass spectrometry on the extracted EV proteins, and performed comparative analysis to identify disease-specific biomarkers. We correlated the levels of these biomarkers with clinical outcome measures and confounding factors. Results The muscle-associated proteins PYGM, MYOM3, FLNC, MYH2 and TTN were exclusively present in DBMD EVs. PYGM, MYOM3, and TTN negatively correlated with age. PYGM and MYOM3 levels were elevated in patients without cardiomyopathy, and PYGM levels were specifically elevated in ambulatory DMD patients. On the other hand, female FSHD patients displayed significantly higher MBL2 and lower GPLD1 levels. However, male FSHD patients exhibited higher C9 and lower C4BPB levels. Additionally, desmosome proteins JUP and DSP were uniquely found in FSHD males. MBL2 positively correlated with age and C4BPB negatively correlated with FSHD severity in male patients. Interpretation Our findings underscore the sensitivity of analyzing circulating EV content to identify disease-specific protein biomarkers for DBMD and FSHD. Our results also emphasize the potential of EV-based biomarker discovery as a promising approach to monitor disease progression as well as effectiveness of therapies in muscular dystrophy, potentially contributing to their approval. Further research with larger cohorts is needed to validate these biomarkers and explore their clinical implications.
Collapse
|
2
|
Moriggi M, Ruggiero L, Torretta E, Zoppi D, Arosio B, Ferri E, Castegna A, Fiorillo C, Gelfi C, Capitanio D. Muscle Proteome Analysis of Facioscapulohumeral Dystrophy Patients Reveals a Metabolic Rewiring Promoting Oxidative/Reductive Stress Contributing to the Loss of Muscle Function. Antioxidants (Basel) 2024; 13:1406. [PMID: 39594549 PMCID: PMC11591206 DOI: 10.3390/antiox13111406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 10/31/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is caused by the epigenetic de-repression of the double homeobox 4 (DUX4) gene, leading to asymmetric muscle weakness and atrophy that begins in the facial and scapular muscles and progresses to the lower limbs. This incurable condition can severely impair muscle function, ultimately resulting in a loss of ambulation. A thorough analysis of molecular factors associated with the varying degrees of muscle impairment in FSHD is still lacking. This study investigates the molecular mechanisms and biomarkers in the biceps brachii of FSHD patients, classified according to the FSHD clinical score, the A-B-C-D classification scheme, and global proteomic variation. Our findings reveal distinct metabolic signatures and compensatory responses in patients. In severe cases, we observe pronounced metabolic dysfunction, marked by dysregulated glycolysis, activation of the reductive pentose phosphate pathway (PPP), a shift toward a reductive TCA cycle, suppression of oxidative phosphorylation, and an overproduction of antioxidants that is not matched by an increase in the redox cofactors needed for their function. This imbalance culminates in reductive stress, exacerbating muscle wasting and inflammation. In contrast, mild cases show metabolic adaptations that mitigate stress by activating polyols and the oxidative PPP, preserving partial energy flow through the oxidative TCA cycle, which supports mitochondrial function and energy balance. Furthermore, activation of the hexosamine biosynthetic pathway promotes autophagy, protecting muscle cells from apoptosis. In conclusion, our proteomic data indicate that specific metabolic alterations characterize both mild and severe FSHD patients. Molecules identified in mild cases may represent potential diagnostic and therapeutic targets for FSHD.
Collapse
Affiliation(s)
- Manuela Moriggi
- Department of Biomedical Sciences for Health, University of Milan, Via Luigi Mangiagalli 31, 20133 Milan, Italy; (M.M.); (D.C.)
| | - Lucia Ruggiero
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples “Federico II”, Via Sergio Pansini 5, 80131 Naples, Italy; (L.R.); (D.Z.)
| | - Enrica Torretta
- Laboratory of Proteomics and Lipidomics, IRCCS Orthopedic Institute Galeazzi, Via R. Galeazzi 4, 20161 Milan, Italy;
| | - Dario Zoppi
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples “Federico II”, Via Sergio Pansini 5, 80131 Naples, Italy; (L.R.); (D.Z.)
| | - Beatrice Arosio
- Department of Clinical Sciences and Community Health, University of Milan, Via della Commenda 19, 20122 Milan, Italy;
| | - Evelyn Ferri
- IRCCS Ca’ Granda Ospedale Maggiore Policlinico Foundation, Via Francesco Sforza 35, 20122 Milan, Italy;
| | - Alessandra Castegna
- Department of Biosciences, Biotechnologies and Environment, University of Bari ALDO MORO, Via Orabona 4, 70125 Bari, Italy;
| | - Chiara Fiorillo
- Child Neuropsychiatric Unit, IRCCS Istituto Giannina Gaslini, DINOGMI-University of Genova, Via Gerolamo Gaslini 5, 16147 Genova, Italy;
| | - Cecilia Gelfi
- Department of Biomedical Sciences for Health, University of Milan, Via Luigi Mangiagalli 31, 20133 Milan, Italy; (M.M.); (D.C.)
- Laboratory of Proteomics and Lipidomics, IRCCS Orthopedic Institute Galeazzi, Via R. Galeazzi 4, 20161 Milan, Italy;
| | - Daniele Capitanio
- Department of Biomedical Sciences for Health, University of Milan, Via Luigi Mangiagalli 31, 20133 Milan, Italy; (M.M.); (D.C.)
| |
Collapse
|
3
|
Vacca V, Rossi C, Pieroni L, De Angelis F, Giacovazzo G, Cicalini I, Ciavardelli D, Pavone F, Coccurello R, Marinelli S. Sex-specific adipose tissue's dynamic role in metabolic and inflammatory response following peripheral nerve injury. iScience 2023; 26:107914. [PMID: 37817933 PMCID: PMC10561049 DOI: 10.1016/j.isci.2023.107914] [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: 05/02/2023] [Revised: 08/30/2023] [Accepted: 09/12/2023] [Indexed: 10/12/2023] Open
Abstract
Epidemiological data and research highlight increased neuropathy and chronic pain prevalence among females, spanning metabolic and normometabolic contexts, including murine models. Prior findings demonstrated diverse immune and neuroimmune responses between genders in neuropathic pain (NeP), alongside distinct protein expression in sciatic nerves. This study unveils adipose tissue's (AT) role in sex-specific NeP responses after peripheral nerve injury. Metabolic assessments, metabolomics, energy expenditure evaluations, AT proteomic analyses, and adipokine mobilization depict distinct AT reactions to nerve damage. Females exhibit altered lipolysis, fatty acid oxidation, heightened energy expenditure, and augmented steroids secretion affecting glucose and insulin metabolism. Conversely, male neuropathy prompts glycolysis, reduced energy expenditure, and lowered unsaturated fatty acid levels. Males' AT promotes regenerative molecules, oxidative stress defense, and stimulates peroxisome proliferator-activated receptors (PPAR-γ) and adiponectin. This study underscores AT's pivotal role in regulating gender-specific inflammatory and metabolic responses to nerve injuries, shedding light on female NeP susceptibility determinants.
Collapse
Affiliation(s)
- Valentina Vacca
- National Council of Research - Institute of Biochemistry and Cell Biology, Monterotondo (RM), Italy
| | - Claudia Rossi
- Department of Innovative Technologies in Medicine and Dentistry, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), "G. D'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Luisa Pieroni
- Departmental Faculty of Medicine, UniCamillus - Saint Camillus International University of Health Sciences, 00131 Rome, Italy
- European Center for Brain Research/Santa Lucia Foundation IRCCS, 00143 Rome, Italy
| | - Federica De Angelis
- National Council of Research - Institute of Biochemistry and Cell Biology, Monterotondo (RM), Italy
- European Center for Brain Research/Santa Lucia Foundation IRCCS, 00143 Rome, Italy
| | - Giacomo Giacovazzo
- European Center for Brain Research/Santa Lucia Foundation IRCCS, 00143 Rome, Italy
- Università degli studi di Teramo (UniTE) - Facoltà di Medicina Veterinaria, 64100 Teramo, Italy
| | - Ilaria Cicalini
- Department of Innovative Technologies in Medicine and Dentistry, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), "G. D'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Domenico Ciavardelli
- Center for Advanced Studies and Technology (CAST), "G. D'Annunzio" University of Chieti-Pescara, Chieti, Italy
- School of Medicine, University Kore of Enna, Enna, Italy
| | - Flaminia Pavone
- National Council of Research - Institute of Biochemistry and Cell Biology, Monterotondo (RM), Italy
| | - Roberto Coccurello
- European Center for Brain Research/Santa Lucia Foundation IRCCS, 00143 Rome, Italy
- Institute for Complex Systems (ISC), National Council of Research (CNR), 00185 Rome, Italy
| | - Sara Marinelli
- National Council of Research - Institute of Biochemistry and Cell Biology, Monterotondo (RM), Italy
| |
Collapse
|
4
|
Nagy K, Gellén G, Papp D, Schlosser G, Révész Á. Optimum collision energies for proteomics: The impact of ion mobility separation. JOURNAL OF MASS SPECTROMETRY : JMS 2023; 58:e4957. [PMID: 37415399 DOI: 10.1002/jms.4957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 05/28/2023] [Accepted: 06/19/2023] [Indexed: 07/08/2023]
Abstract
Ion mobility spectrometry (IMS) is a widespread separation technique used in various research fields. It can be coupled to liquid chromatography-mass spectrometry (LC-MS/MS) methods providing an additional separation dimension. During IMS, ions are subjected to multiple collisions with buffer gas, which may cause significant ion heating. The present project addresses this phenomenon from the bottom-up proteomics point of view. We performed LC-MS/MS measurements on a cyclic ion mobility mass spectrometer with varied collision energy (CE) settings both with and without IMS. We investigated the CE dependence of identification score, using Byonic search engine, for more than 1000 tryptic peptides from HeLa digest standard. We determined the optimal CE values-giving the highest identification score-for both setups (i.e., with and without IMS). Results show that lower CE is advantageous when IMS separation is applied, by 6.3 V on average. This value belongs to the one-cycle separation configuration, and multiple cycles may supposedly have even larger impact. The effect of IMS is also reflected in the trends of optimal CE values versus m/z functions. The parameters suggested by the manufacturer were found to be almost optimal for the setup without IMS; on the other hand, they are obviously too high with IMS. Practical consideration on setting up a mass spectrometric platform hyphenated to IMS is also presented. Furthermore, the two CID (collision induced dissociation) fragmentation cells of the instrument-located before and after the IMS cell-were also compared, and we found that CE adjustment is needed when the trap cell is used for activation instead of the transfer cell. Data have been deposited in the MassIVE repository (MSV000090944).
Collapse
Affiliation(s)
- Kinga Nagy
- MS Proteomics Research Group, Research Centre for Natural Sciences, Budapest, H-1117, Hungary
- Hevesy György PhD School of Chemistry, Faculty of Science, Institute of Chemistry, Eötvös Loránd University, Budapest, H-1117, Hungary
| | - Gabriella Gellén
- MTA-ELTE Lendület Ion Mobility Mass Spectrometry Research Group, Faculty of Science, Institute of Chemistry, Eötvös Loránd University, Budapest, H-1117, Hungary
| | - Dávid Papp
- Hevesy György PhD School of Chemistry, Faculty of Science, Institute of Chemistry, Eötvös Loránd University, Budapest, H-1117, Hungary
- MTA-ELTE Lendület Ion Mobility Mass Spectrometry Research Group, Faculty of Science, Institute of Chemistry, Eötvös Loránd University, Budapest, H-1117, Hungary
| | - Gitta Schlosser
- MTA-ELTE Lendület Ion Mobility Mass Spectrometry Research Group, Faculty of Science, Institute of Chemistry, Eötvös Loránd University, Budapest, H-1117, Hungary
| | - Ágnes Révész
- MS Proteomics Research Group, Research Centre for Natural Sciences, Budapest, H-1117, Hungary
| |
Collapse
|
5
|
Strafella C, Caputo V, Bortolani S, Torchia E, Megalizzi D, Trastulli G, Monforte M, Colantoni L, Caltagirone C, Ricci E, Tasca G, Cascella R, Giardina E. Whole exome sequencing highlights rare variants in CTCF, DNMT1, DNMT3A, EZH2 and SUV39H1 as associated with FSHD. Front Genet 2023; 14:1235589. [PMID: 37674478 PMCID: PMC10477786 DOI: 10.3389/fgene.2023.1235589] [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: 06/06/2023] [Accepted: 08/09/2023] [Indexed: 09/08/2023] Open
Abstract
Introduction: Despite the progress made in the study of Facioscapulohumeral Dystrophy (FSHD), the wide heterogeneity of disease complicates its diagnosis and the genotype-phenotype correlation among patients and within families. In this context, the present work employed Whole Exome Sequencing (WES) to investigate known and unknown genetic contributors that may be involved in FSHD and may represent potential disease modifiers, even in presence of a D4Z4 Reduced Allele (DRA). Methods: A cohort of 126 patients with clinical signs of FSHD were included in the study, which were characterized by D4Z4 sizing, methylation analysis and WES. Specific protocols were employed for D4Z4 sizing and methylation analysis, whereas the Illumina® Next-Seq 550 system was utilized for WES. The study included both patients with a DRA compatible with FSHD diagnosis and patients with longer D4Z4 alleles. In case of patients harboring relevant variants from WES, the molecular analysis was extended to the family members. Results: The WES data analysis highlighted 20 relevant variants, among which 14 were located in known genetic modifiers (SMCHD1, DNMT3B and LRIF1) and 6 in candidate genes (CTCF, DNMT1, DNMT3A, EZH2 and SUV39H1). Most of them were found together with a permissive short (4-7 RU) or borderline/long DRA (8-20 RU), supporting the possibility that different genes can contribute to disease heterogeneity in presence of a FSHD permissive background. The segregation and methylation analysis among family members, together with clinical findings, provided a more comprehensive picture of patients. Discussion: Our results support FSHD pathomechanism being complex with a multigenic contribution by several known (SMCHD1, DNMT3B, LRIF1) and possibly other candidate genes (CTCF, DNMT1, DNMT3A, EZH2, SUV39H1) to disease penetrance and expressivity. Our results further emphasize the importance of extending the analysis of molecular findings within the proband's family, with the purpose of providing a broader framework for understanding single cases and allowing finer genotype-phenotype correlations in FSHD-affected families.
Collapse
Affiliation(s)
- Claudia Strafella
- Genomic Medicine Laboratory UILDM, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Valerio Caputo
- Genomic Medicine Laboratory UILDM, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Sara Bortolani
- Unità Operativa Complessa di Neurologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Eleonora Torchia
- Unità Operativa Complessa di Neurologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Domenica Megalizzi
- Genomic Medicine Laboratory UILDM, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Giulia Trastulli
- Genomic Medicine Laboratory UILDM, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Mauro Monforte
- Unità Operativa Complessa di Neurologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Luca Colantoni
- Genomic Medicine Laboratory UILDM, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Carlo Caltagirone
- Department of Clinical and Behavioral Neurology, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Enzo Ricci
- Unità Operativa Complessa di Neurologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Istituto di Neurologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giorgio Tasca
- Unità Operativa Complessa di Neurologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle UponTyne, United Kingdom
| | - Raffaella Cascella
- Genomic Medicine Laboratory UILDM, IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Biomedical Sciences, Catholic University Our Lady of Good Counsel, Tirana, Albania
| | - Emiliano Giardina
- Genomic Medicine Laboratory UILDM, IRCCS Santa Lucia Foundation, Rome, Italy
- Medical Genetics Laboratory, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| |
Collapse
|
6
|
Banerji CRS, Greco A, Joosten LAB, van Engelen BGM, Zammit PS. The FSHD muscle-blood biomarker: a circulating transcriptomic biomarker for clinical severity in facioscapulohumeral muscular dystrophy. Brain Commun 2023; 5:fcad221. [PMID: 37731904 PMCID: PMC10507741 DOI: 10.1093/braincomms/fcad221] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 06/20/2023] [Accepted: 08/14/2023] [Indexed: 09/22/2023] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is a prevalent, incurable skeletal myopathy. Clinical trials for FSHD are hindered by heterogeneous biomarkers poorly associated with clinical severity, requiring invasive muscle biopsy. Macroscopically, FSHD presents with slow fatty replacement of muscle, rapidly accelerated by inflammation. Mis-expression of the transcription factor DUX4 is currently accepted to underlie pathogenesis, and mechanisms including PAX7 target gene repression have been proposed. Here, we performed RNA-sequencing on MRI-guided inflamed and isogenic non-inflamed muscle biopsies from the same clinically characterized FSHD patients (n = 24), alongside isogenic peripheral blood mononucleated cells from a subset of patients (n = 13) and unaffected controls (n = 11). Multivariate models were employed to evaluate the clinical associations of five published FSHD transcriptomic biomarkers. We demonstrated that PAX7 target gene repression can discriminate control, inflamed and non-inflamed FSHD muscle independently of age and sex (P < 0.013), while the discriminatory power of DUX4 target genes was limited to distinguishing FSHD muscle from control. Importantly, the level of PAX7 target gene repression in non-inflamed muscle associated with clinical assessments of FSHD severity (P = 0.04). DUX4 target gene biomarkers in FSHD muscle showed associations with lower limb fat fraction and D4Z4 array length but not clinical assessment. Lastly, PAX7 target gene repression in FSHD muscle correlated with the level in isogenic peripheral blood mononucleated cells (P = 0.002). A refined PAX7 target gene biomarker comprising 143/601 PAX7 target genes computed in peripheral blood (the FSHD muscle-blood biomarker) associated with clinical severity in FSHD patients (P < 0.036). Our new circulating biomarker validates as a classifier of clinical severity in an independent data set of 54 FSHD patient and 29 matched control blood samples, with improved power in older patients (P = 0.03). In summary, we present the minimally invasive FSHD muscle-blood biomarker of FSHD clinical severity valid in patient muscle and blood, of potential use in routine disease monitoring and clinical trials.
Collapse
Affiliation(s)
- Christopher R S Banerji
- Randall Centre for Cell and Molecular Biophysics, King’s College London, London SE1 1UL, UK
- The Alan Turing Institute, The British Library, London NW1 2DB, UK
| | - Anna Greco
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
- Department of Internal Medicine, Radboud Institute of Molecular Life Sciences (RIMLS) and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen 6525, The Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud Institute of Molecular Life Sciences (RIMLS) and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen 6525, The Netherlands
- Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Baziel G M van Engelen
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Peter S Zammit
- Randall Centre for Cell and Molecular Biophysics, King’s College London, London SE1 1UL, UK
| |
Collapse
|
7
|
Nishimura Y, Bittel AJ, Stead CA, Chen YW, Burniston JG. Facioscapulohumeral Muscular Dystrophy is Associated With Altered Myoblast Proteome Dynamics. Mol Cell Proteomics 2023; 22:100605. [PMID: 37353005 PMCID: PMC10392138 DOI: 10.1016/j.mcpro.2023.100605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 05/31/2023] [Accepted: 06/20/2023] [Indexed: 06/25/2023] Open
Abstract
Proteomic studies in facioscapulohumeral muscular dystrophy (FSHD) could offer new insight into disease mechanisms underpinned by post-transcriptional processes. We used stable isotope (deuterium oxide; D2O) labeling and peptide mass spectrometry to investigate the abundance and turnover rates of proteins in cultured muscle cells from two individuals affected by FSHD and their unaffected siblings (UASb). We measured the abundance of 4420 proteins and the turnover rate of 2324 proteins in each (n = 4) myoblast sample. FSHD myoblasts exhibited a greater abundance but slower turnover rate of subunits of mitochondrial respiratory complexes and mitochondrial ribosomal proteins, which may indicate an accumulation of "older" less viable mitochondrial proteins in myoblasts from individuals affected by FSHD. Treatment with a 2'-O-methoxyethyl modified antisense oligonucleotide targeting exon 3 of the double homeobox 4 (DUX4) transcript tended to reverse mitochondrial protein dysregulation in FSHD myoblasts, indicating the effect on mitochondrial proteins may be a DUX4-dependent mechanism. Our results highlight the importance of post-transcriptional processes and protein turnover in FSHD pathology and provide a resource for the FSHD research community to explore this burgeoning aspect of FSHD.
Collapse
Affiliation(s)
- Yusuke Nishimura
- Research Institute for Sport & Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Adam J Bittel
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, District of Columbia, USA
| | - Connor A Stead
- Research Institute for Sport & Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Yi-Wen Chen
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, District of Columbia, USA.
| | - Jatin G Burniston
- Research Institute for Sport & Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom.
| |
Collapse
|
8
|
Montagnese F, de Valle K, Lemmers RJLF, Mul K, Dumonceaux J, Voermans N. 268th ENMC workshop - Genetic diagnosis, clinical classification, outcome measures, and biomarkers in Facioscapulohumeral Muscular Dystrophy (FSHD): Relevance for clinical trials. Neuromuscul Disord 2023; 33:447-462. [PMID: 37099914 DOI: 10.1016/j.nmd.2023.04.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 04/07/2023] [Indexed: 04/28/2023]
Affiliation(s)
- Federica Montagnese
- Department of Neurology, Ludwig-Maximilian University Munich, Friedrich-Baur-Institute, Germany
| | - Katy de Valle
- Department of Neurology, The Royal Children's Hospital, Melbourne, Australia
| | - Richard J L F Lemmers
- Department Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Karlien Mul
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Julie Dumonceaux
- NIHR Biomedical Research Centre, University College London, Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust, United Kingdom of Great Britain and Northern Ireland, London WC1N 1EH, United Kingdom
| | - Nicol Voermans
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
9
|
Claus C, Slavin M, Ansseau E, Lancelot C, Bah K, Lassche S, Fiévet M, Greco A, Tomaiuolo S, Tassin A, Dudome V, Kusters B, Declèves AE, Laoudj-Chenivesse D, van Engelen BGM, Nonclercq D, Belayew A, Kalisman N, Coppée F. The double homeodomain protein DUX4c is associated with regenerating muscle fibers and RNA-binding proteins. Skelet Muscle 2023; 13:5. [PMID: 36882853 PMCID: PMC9990282 DOI: 10.1186/s13395-022-00310-y] [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: 07/20/2021] [Accepted: 11/30/2022] [Indexed: 03/09/2023] Open
Abstract
BACKGROUND We have previously demonstrated that double homeobox 4 centromeric (DUX4C) encoded for a functional DUX4c protein upregulated in dystrophic skeletal muscles. Based on gain- and loss-of-function studies we have proposed DUX4c involvement in muscle regeneration. Here, we provide further evidence for such a role in skeletal muscles from patients affected with facioscapulohumeral muscular dystrophy (FSHD). METHODS DUX4c was studied at RNA and protein levels in FSHD muscle cell cultures and biopsies. Its protein partners were co-purified and identified by mass spectrometry. Endogenous DUX4c was detected in FSHD muscle sections with either its partners or regeneration markers using co-immunofluorescence or in situ proximity ligation assay. RESULTS We identified new alternatively spliced DUX4C transcripts and confirmed DUX4c immunodetection in rare FSHD muscle cells in primary culture. DUX4c was detected in nuclei, cytoplasm or at cell-cell contacts between myocytes and interacted sporadically with specific RNA-binding proteins involved, a.o., in muscle differentiation, repair, and mass maintenance. In FSHD muscle sections, DUX4c was found in fibers with unusual shape or central/delocalized nuclei (a regeneration feature) staining for developmental myosin heavy chain, MYOD or presenting intense desmin labeling. Some couples of myocytes/fibers locally exhibited peripheral DUX4c-positive areas that were very close to each other, but in distinct cells. MYOD or intense desmin staining at these locations suggested an imminent muscle cell fusion. We further demonstrated DUX4c interaction with its major protein partner, C1qBP, inside myocytes/myofibers that presented features of regeneration. On adjacent muscle sections, we could unexpectedly detect DUX4 (the FSHD causal protein) and its interaction with C1qBP in fusing myocytes/fibers. CONCLUSIONS DUX4c upregulation in FSHD muscles suggests it contributes not only to the pathology but also, based on its protein partners and specific markers, to attempts at muscle regeneration. The presence of both DUX4 and DUX4c in regenerating FSHD muscle cells suggests DUX4 could compete with normal DUX4c functions, thus explaining why skeletal muscle is particularly sensitive to DUX4 toxicity. Caution should be exerted with therapeutic agents aiming for DUX4 suppression because they might also repress the highly similar DUX4c and interfere with its physiological role.
Collapse
Affiliation(s)
- Clothilde Claus
- Laboratory of Metabolic and Molecular Biochemistry, Research Institute for Health Sciences and Technology, University of Mons, 6, Avenue du Champs de Mars, B-7000, Mons, Belgium
| | - Moriya Slavin
- Department of Biological Chemistry, the Alexander Silberman Institute of Life Sciences, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Eugénie Ansseau
- Laboratory of Metabolic and Molecular Biochemistry, Research Institute for Health Sciences and Technology, University of Mons, 6, Avenue du Champs de Mars, B-7000, Mons, Belgium
| | - Céline Lancelot
- Laboratory of Metabolic and Molecular Biochemistry, Research Institute for Health Sciences and Technology, University of Mons, 6, Avenue du Champs de Mars, B-7000, Mons, Belgium
| | - Karimatou Bah
- Laboratory of Metabolic and Molecular Biochemistry, Research Institute for Health Sciences and Technology, University of Mons, 6, Avenue du Champs de Mars, B-7000, Mons, Belgium
| | - Saskia Lassche
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands.,Department of Neurology, Zuyderland Medical Center, Heerlen, the Netherlands
| | - Manon Fiévet
- Laboratory of Metabolic and Molecular Biochemistry, Research Institute for Health Sciences and Technology, University of Mons, 6, Avenue du Champs de Mars, B-7000, Mons, Belgium
| | - Anna Greco
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | - Sara Tomaiuolo
- Laboratory of Metabolic and Molecular Biochemistry, Research Institute for Health Sciences and Technology, University of Mons, 6, Avenue du Champs de Mars, B-7000, Mons, Belgium
| | - Alexandra Tassin
- Laboratory of Metabolic and Molecular Biochemistry, Research Institute for Health Sciences and Technology, University of Mons, 6, Avenue du Champs de Mars, B-7000, Mons, Belgium.,Laboratory of Respiratory Physiology and Rehabilitation, Research Institute for Health Sciences and Technology, University of Mons, 6, Avenue du Champs de Mars, B-7000, Mons, Belgium
| | - Virginie Dudome
- Laboratory of Metabolic and Molecular Biochemistry, Research Institute for Health Sciences and Technology, University of Mons, 6, Avenue du Champs de Mars, B-7000, Mons, Belgium
| | - Benno Kusters
- Department of Pathology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | - Anne-Emilie Declèves
- Laboratory of Metabolic and Molecular Biochemistry, Research Institute for Health Sciences and Technology, University of Mons, 6, Avenue du Champs de Mars, B-7000, Mons, Belgium
| | | | - Baziel G M van Engelen
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | - Denis Nonclercq
- Laboratory of Histology, Research Institute for Health Sciences and Technology, University of Mons, 6, Avenue du Champs de Mars, B-7000, Mons, Belgium
| | - Alexandra Belayew
- Laboratory of Metabolic and Molecular Biochemistry, Research Institute for Health Sciences and Technology, University of Mons, 6, Avenue du Champs de Mars, B-7000, Mons, Belgium
| | - Nir Kalisman
- Department of Biological Chemistry, the Alexander Silberman Institute of Life Sciences, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Frédérique Coppée
- Laboratory of Metabolic and Molecular Biochemistry, Research Institute for Health Sciences and Technology, University of Mons, 6, Avenue du Champs de Mars, B-7000, Mons, Belgium.
| |
Collapse
|
10
|
Tihaya MS, Mul K, Balog J, de Greef JC, Tapscott SJ, Tawil R, Statland JM, van der Maarel SM. Facioscapulohumeral muscular dystrophy: the road to targeted therapies. Nat Rev Neurol 2023; 19:91-108. [PMID: 36627512 PMCID: PMC11578282 DOI: 10.1038/s41582-022-00762-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2022] [Indexed: 01/11/2023]
Abstract
Advances in the molecular understanding of facioscapulohumeral muscular dystrophy (FSHD) have revealed that FSHD results from epigenetic de-repression of the DUX4 gene in skeletal muscle, which encodes a transcription factor that is active in early embryonic development but is normally silenced in almost all somatic tissues. These advances also led to the identification of targets for disease-altering therapies for FSHD, as well as an improved understanding of the molecular mechanism of the disease and factors that influence its progression. Together, these developments led the FSHD research community to shift its focus towards the development of disease-modifying treatments for FSHD. This Review presents advances in the molecular and clinical understanding of FSHD, discusses the potential targeted therapies that are currently being explored, some of which are already in clinical trials, and describes progress in the development of FSHD-specific outcome measures and assessment tools for use in future clinical trials.
Collapse
Affiliation(s)
- Mara S Tihaya
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Karlien Mul
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Judit Balog
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Jessica C de Greef
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Stephen J Tapscott
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Rabi Tawil
- Department of Neurology, University of Rochester Medical Center, Rochester, NY, USA
| | - Jeffrey M Statland
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | | |
Collapse
|
11
|
Monforte M, Attarian S, Vissing J, Diaz-Manera J, Tasca G. 265th ENMC International Workshop: Muscle imaging in Facioscapulohumeral Muscular Dystrophy (FSHD): relevance for clinical trials. 22-24 April 2022, Hoofddorp, The Netherlands. Neuromuscul Disord 2023; 33:65-75. [PMID: 36369218 DOI: 10.1016/j.nmd.2022.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/10/2022] [Accepted: 10/19/2022] [Indexed: 11/07/2022]
Affiliation(s)
- Mauro Monforte
- Unità Operativa Complessa di Neurologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Shahram Attarian
- Reference Center for Neuromuscular Disorders and ALS, CHU La Timone Aix-Marseille Hospital University Marseille, France
| | - John Vissing
- Copenhagen Neuromuscular Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Jordi Diaz-Manera
- John Walton Muscular Dystrophy Research Center, University of Newcastle, Newcastle upon Tyne, United Kingdom
| | - Giorgio Tasca
- Unità Operativa Complessa di Neurologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, Rome 00168, Italy.
| |
Collapse
|
12
|
Campbell AE, Arjomand J, King OD, Tawil R, Jagannathan S. A Targeted Approach for Evaluating DUX4-Regulated Proteins as Potential Serum Biomarkers for Facioscapulohumeral Muscular Dystrophy Using Immunoassay Proteomics. J Neuromuscul Dis 2023; 10:1031-1040. [PMID: 37899061 PMCID: PMC10657687 DOI: 10.3233/jnd-221636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/28/2023] [Indexed: 10/31/2023]
Abstract
BACKGROUND Facioscapulohumeral muscular dystrophy (FSHD) is a progressive myopathy caused by misexpression of the double homeobox 4 (DUX4) embryonic transcription factor in skeletal muscle. Identifying quantitative and minimally invasive FSHD biomarkers to report on DUX4 activity will significantly accelerate therapeutic development. OBJECTIVE The goal of this study was to analyze secreted proteins known to be induced by DUX4 using the commercially available Olink Proteomics platform in order to identify potential blood-based molecular FSHD biomarkers. METHODS We used high-throughput, multiplex immunoassays from Olink Proteomics to measure the levels of several known DUX4-induced genes in a cellular myoblast model of FSHD, in FSHD patient-derived myotube cell cultures, and in serum from individuals with FSHD. Levels of other proteins on the Olink Proteomics panels containing these DUX4 targets were also examined in secondary exploratory analysis. RESULTS Placental alkaline phosphatase (ALPP) levels correlated with DUX4 expression in both cell-based FSHD systems but did not distinguish FSHD patient serum from unaffected controls. CONCLUSIONS ALPP, as measured with the Olink Proteomics platform, is not a promising FSHD serum biomarker candidate but could be utilized to evaluate DUX4 activity in discovery research efforts.
Collapse
Affiliation(s)
- Amy E. Campbell
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Oliver D. King
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Rabi Tawil
- Department of Neurology, University of Rochester Medical Center, Rochester, NY, USA
| | - Sujatha Jagannathan
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- RNA Bioscience Initiative, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
13
|
Wong CJ, Wang L, Holers VM, Frazer-Abel A, van der Maarel SM, Tawil R, Statland JM, Tapscott SJ. Elevated plasma complement components in facioscapulohumeral dystrophy. Hum Mol Genet 2022; 31:1821-1829. [PMID: 34919696 PMCID: PMC9169453 DOI: 10.1093/hmg/ddab364] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/07/2021] [Accepted: 12/14/2021] [Indexed: 11/12/2022] Open
Abstract
Advances in understanding the pathophysiology of facioscapulohumeral dystrophy (FSHD) have led to several therapeutic approaches entering clinical trials and an increased need to develop biomarkers of disease activity and progression. Multiple prior studies have shown early elevation of RNAs encoding components of the complement pathways and relatively widespread activated complement complexes by immunodetection in FSHD muscle. The current study tested plasma from two independent cohorts of FSHD and control subjects and found elevated complement components in both FSHD cohorts. Combining subjects from both cohorts identified complement factors that best distinguished FSHD and controls. Within the FSHD group, a subset of subjects showed elevation in multiple complement components. Together these findings suggest the need for future studies to determine whether measurements of complement activation can be used as a non-invasive measurement of FSHD disease activity, progression and/or response to therapies. In addition, with the ongoing expansion of complement therapeutic approaches, consideration for precision-based targeting of this pathway is appropriate.
Collapse
Affiliation(s)
- Chao-Jen Wong
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Leo Wang
- Department of Neurology, University of Washington, Seattle, WA 98105, USA
| | - V Michael Holers
- Division of Rheumatology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Ashley Frazer-Abel
- Exsera BioLabs, Division of Rheumatalogy, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | | | - Rabi Tawil
- Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Jeffrey M Statland
- Department of Neurology, University of Kansas Medical Center, Kansas City, KA 66160, USA
| | - Stephen J Tapscott
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Department of Neurology, University of Washington, Seattle, WA 98105, USA
| | | |
Collapse
|
14
|
Militello R, Pinto G, Illiano A, Luti S, Magherini F, Amoresano A, Modesti PA, Modesti A. Modulation of Plasma Proteomic Profile by Regular Training in Male and Female Basketball Players: A Preliminary Study. Front Physiol 2022; 13:813447. [PMID: 35360242 PMCID: PMC8964093 DOI: 10.3389/fphys.2022.813447] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 01/20/2022] [Indexed: 11/13/2022] Open
Abstract
Monitoring fatigue and recovery during training periods contributes to identifying the best training methods to achieve sports performance. To date, little is known about sex-related differences in sports adaptations. The aim of the present study is to identify sex-related sports adaptation proteins in female basketball players and male basketball players using proteomics approach on plasma samples withdrawn from athletes during in-season training period but far from a competition. A cohort of 20 professional basketball players, 10 female (BF) and 10 male (BM), and 20 sedentary male (10 CM) and female (10 CF) as control, of comparable age and BMI, were involved in this study. Protein profiles of plasma samples obtained from BM, BF, CM, and CF were analyzed by two-dimensional electrophoresis (2-DE). Differentially expressed proteins were identified by mass spectrometry. The computational 2-DE gel image analysis pointed out 33 differentially expressed protein spots (ANOVA p-value < 0.05) and differences between male and female basketball players are more evident among the players than controls. The expression profile of 54.5% of the total proteins is affected by sports activity. Furthermore, 14 proteins are differentially expressed in basket female players in comparison with their relative controls while seven are differentially expressed in basket male players in comparison with their controls. In conclusion, we identify in female athletes a reduction in proteins related to transcription regulation, most of these modulate chronic inflammation confirming the anti-inflammatory effect of regular training in female muscle metabolism. In male and female athletes, we found a decrease in Transthyretin involved in muscle homeostasis and regeneration and Dermcidin a stress-induced myokine linked to inflammatory and it will be interesting to fully understand the role of its different isoforms in male and female skeletal muscle contraction.
Collapse
Affiliation(s)
- Rosamaria Militello
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio," University of Florence, Florence, Italy
| | - Gabriella Pinto
- Istituto Nazionale Biostrutture e Biosistemi, Rome, Italy.,Department of Chemical Sciences, Polytechnic and Basic Sciences School, University of Naples Federico II, Naples, Italy
| | - Anna Illiano
- Istituto Nazionale Biostrutture e Biosistemi, Rome, Italy.,Department of Chemical Sciences, Polytechnic and Basic Sciences School, University of Naples Federico II, Naples, Italy
| | - Simone Luti
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio," University of Florence, Florence, Italy
| | - Francesca Magherini
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio," University of Florence, Florence, Italy
| | | | - Pietro Amedeo Modesti
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alessandra Modesti
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio," University of Florence, Florence, Italy
| |
Collapse
|
15
|
Laberthonnière C, Novoa-Del-Toro EM, Delourme M, Chevalier R, Broucqsault N, Mazaleyrat K, Streichenberger N, Manel V, Bernard R, Salort Campana E, Attarian S, Nguyen K, Robin JD, Baudot A, Magdinier F. Facioscapulohumeral dystrophy weakened sarcomeric contractility is mimicked in induced pluripotent stem cells-derived innervated muscle fibres. J Cachexia Sarcopenia Muscle 2022; 13:621-635. [PMID: 34859613 PMCID: PMC8818656 DOI: 10.1002/jcsm.12835] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 09/14/2021] [Accepted: 09/20/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Facioscapulohumeral dystrophy (FSHD) is a late-onset autosomal dominant form of muscular dystrophy involving specific groups of muscles with variable weakness that precedes inflammatory response, fat infiltration, and muscle atrophy. As there is currently no cure for this disease, understanding and modelling the typical muscle weakness in FSHD remains a major milestone towards deciphering the disease pathogenesis as it will pave the way to therapeutic strategies aimed at correcting the functional muscular defect in patients. METHODS To gain further insights into the specificity of the muscle alteration in this disease, we derived induced pluripotent stem cells from patients affected with Types 1 and 2 FSHD but also from patients affected with Bosma arhinia and microphthalmia. We differentiated these cells into contractile innervated muscle fibres and analysed their transcriptome by RNA Seq in comparison with cells derived from healthy donors. To uncover biological pathways altered in the disease, we applied MOGAMUN, a multi-objective genetic algorithm that integrates multiplex complex networks of biological interactions (protein-protein interactions, co-expression, and biological pathways) and RNA Seq expression data to identify active modules. RESULTS We identified 132 differentially expressed genes that are specific to FSHD cells (false discovery rate < 0.05). In FSHD, the vast majority of active modules retrieved with MOGAMUN converges towards a decreased expression of genes encoding proteins involved in sarcomere organization (P value 2.63e-12 ), actin cytoskeleton (P value 9.4e-5 ), myofibril (P value 2.19e-12 ), actin-myosin sliding, and calcium handling (with P values ranging from 7.9e-35 to 7.9e-21 ). Combined with in vivo validations and functional investigations, our data emphasize a reduction in fibre contraction (P value < 0.0001) indicating that the muscle weakness that is typical of FSHD clinical spectrum might be associated with dysfunction of calcium release (P value < 0.0001), actin-myosin interactions, motor activity, mechano-transduction, and dysfunctional sarcomere contractility. CONCLUSIONS Identification of biomarkers of FSHD muscle remain critical for understanding the process leading to the pathology but also for the definition of readouts to be used for drug design, outcome measures, and monitoring of therapies. The different pathways identified through a system biology approach have been largely overlooked in the disease. Overall, our work opens new perspectives in the definition of biomarkers able to define the muscle alteration but also in the development of novel strategies to improve muscle function as it provides functional parameters for active molecule screening.
Collapse
Affiliation(s)
| | | | - Mégane Delourme
- Aix Marseille Univ, INSERM, Marseille Medical Genetics, MMG, Marseille, France
| | - Raphaël Chevalier
- Aix Marseille Univ, INSERM, Marseille Medical Genetics, MMG, Marseille, France
| | - Natacha Broucqsault
- Aix Marseille Univ, INSERM, Marseille Medical Genetics, MMG, Marseille, France
| | - Kilian Mazaleyrat
- Aix Marseille Univ, INSERM, Marseille Medical Genetics, MMG, Marseille, France
| | - Nathalie Streichenberger
- Neuropathology, Lyon Civil Hospices, Lyon, France.,Neuromyogène Institute, CNRS-UMR 5310, INSERM 1217, Claude Bernard University Lyon 1, University of Lyon, Lyon, France.,Reference Centre for Neuromuscular Diseases, Hospital for Woman Mother Child, Lyon, France
| | - Véronique Manel
- Department of Medical Genetics, Timone Infant Hospital, Marseille, France
| | - Rafaëlle Bernard
- Aix Marseille Univ, INSERM, Marseille Medical Genetics, MMG, Marseille, France.,Reference Centre for Neuromuscular Diseases and ALS, Timone Adult Hospital, Marseille, France
| | | | - Shahram Attarian
- Aix Marseille Univ, INSERM, Marseille Medical Genetics, MMG, Marseille, France
| | - Karine Nguyen
- Aix Marseille Univ, INSERM, Marseille Medical Genetics, MMG, Marseille, France.,Reference Centre for Neuromuscular Diseases and ALS, Timone Adult Hospital, Marseille, France
| | - Jérôme D Robin
- Aix Marseille Univ, INSERM, Marseille Medical Genetics, MMG, Marseille, France
| | - Anais Baudot
- Aix Marseille Univ, INSERM, Marseille Medical Genetics, MMG, Marseille, France
| | | |
Collapse
|
16
|
Carregari VC. Protein Extraction and Sample Preparation Methods for Shotgun Proteomics with Central Nervous System Cells and Brain Tissue. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1382:1-15. [DOI: 10.1007/978-3-031-05460-0_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
17
|
Functional Identification of Complement Factor D and Analysis of Its Expression during GCRV Infection in Grass Carp ( Ctenopharyngodon idella). Int J Mol Sci 2021; 22:ijms222112011. [PMID: 34769442 PMCID: PMC8584590 DOI: 10.3390/ijms222112011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/19/2021] [Accepted: 11/02/2021] [Indexed: 12/12/2022] Open
Abstract
Complement factor D (Df) is a serine protease well known for activating the alternative pathway (AP) in mammals by promoting the cleavage of complement component 3 (C3), thus becoming involved in innate defense. In teleost fish, however, the functional mechanisms of Df in the AP and against pathogen infection are far from clear. In the present study, we cloned and characterized the Df gene, CiDf, from grass carp (Ctenopharyngodon idella) and analyzed its function in promoting C3 cleavage and expression changes after grass carp reovirus (GCRV) infection. The open reading frame of CiDf was found to be 753 bp, encoding 250 amino acids with a molecular mass of 27.06 kDa. CiDf harbors a conserved Tryp_SPc domain, with three conserved residues representing the catalytic triad and three conserved binding sites in the substrate specificity pocket. Pairwise alignment showed that CiDf shares the highest identity (96%) and similarity (98%) with Df from Anabarilius grahami. Phylogenetic analysis indicated that CiDf and other fish Dfs formed a distinct evolutionary branch. Similar to most Dfs from other vertebrates, the CiDf gene structure is characterized by four introns and five exons. The incubation of recombinant CiDf protein with grass carp serum significantly increased the C3b content, demonstrating the conserved function of CiDf in the AP in promoting C3 cleavage, similar to Dfs in mammals. CiDf mRNA expression was widely detected in various tissues and levels were relatively higher in the liver, spleen, and intestine of grass carp. During GCRV infection over a 168-hour period, a high level of CiDf mRNA expression in the liver, spleen, and intestine was maintained at 144 and 168 h, suggesting AP activity at the late stage of GCRV infection. Collectively, the above results reveal the conserved structure and function of CiDf and its distinct expression patterns after GCRV infection, which provide a key basis for studying the roles of Df and AP during GCRV infection in the grass carp C. idella.
Collapse
|
18
|
Monforte M, Bortolani S, Torchia E, Cristiano L, Laschena F, Tartaglione T, Ricci E, Tasca G. Diagnostic magnetic resonance imaging biomarkers for facioscapulohumeral muscular dystrophy identified by machine learning. J Neurol 2021; 269:2055-2063. [PMID: 34486074 DOI: 10.1007/s00415-021-10786-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/29/2021] [Accepted: 08/30/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND The diagnosis of facioscapulohumeral muscular dystrophy (FSHD) can be challenging in patients not displaying the classical phenotype or with atypical clinical features. Despite the identification by magnetic resonance imaging (MRI) of selective patterns of muscle involvement, their specificity and added diagnostic value are unknown. METHODS We aimed to identify the radiological features more useful to distinguish FSHD from other myopathies and test the diagnostic accuracy of MRI. A retrospective cohort of 295 patients (187 FSHD, 108 non-FSHD) studied by upper and lower-limb muscle MRI was analyzed. Scans were evaluated for the presence of 15 radiological features. A random forest machine learning algorithm was used to identify the most relevant for FSHD diagnosis. Different patterns were created by their combination and diagnostic accuracy of each of them was tested. RESULTS The combination of trapezius involvement and bilateral subscapularis muscle sparing achieved the best diagnostic accuracy (0.89, 95% Confidence Interval [0.85-0.92]) with 0.90 [0.85-0.94] sensitivity and 0.88 [0.80-0.93] specificity. This pattern correctly identified 91% atypical FSHD patients of our cohort. The combination of trapezius involvement, bilateral subscapularis and iliopsoas sparing and asymmetric involvement of upper and lower-limb muscles was pathognomonic for FSHD, yielding a specificity of 0.99 [0.95-1.00]. CONCLUSIONS We identified MRI patterns that showed a high diagnostic power in promptly discriminating FSHD from other muscle disorders, with comparable performance irrespective of typical or atypical clinical features. Upper girdle in addition to lower-limb muscle imaging should be extensively implemented in the diagnostic workup to support or exclude a diagnosis of FSHD.
Collapse
Affiliation(s)
- Mauro Monforte
- Unità Operativa Complessa di Neurologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168, Rome, Italy.
| | - Sara Bortolani
- Unità Operativa Complessa di Neurologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168, Rome, Italy
| | - Eleonora Torchia
- Unità Operativa Complessa di Neurologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168, Rome, Italy
| | | | | | - Tommaso Tartaglione
- Dipartimento di Radiologia, IDI IRCCS, Rome, Italy.,Istituto di Radiologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Enzo Ricci
- Unità Operativa Complessa di Neurologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168, Rome, Italy. .,Istituto di Neurologia, Università Cattolica del Sacro Cuore, Rome, Italy.
| | - Giorgio Tasca
- Unità Operativa Complessa di Neurologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168, Rome, Italy
| |
Collapse
|
19
|
Voermans NC, Vriens-Munoz Bravo M, Padberg GW, Laforêt P. 1st FSHD European Trial Network workshop:Working towards trial readiness across Europe. Neuromuscul Disord 2021; 31:907-918. [PMID: 34404575 DOI: 10.1016/j.nmd.2021.07.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 07/15/2021] [Indexed: 01/29/2023]
Affiliation(s)
- N C Voermans
- FSHD Europe, Radboud University Medical Centre, P.O. Box 9101, Nijmegen 6500 HB, the Netherlands.
| | - M Vriens-Munoz Bravo
- FSHD Europe, Radboud University Medical Centre, P.O. Box 9101, Nijmegen 6500 HB, the Netherlands
| | - G W Padberg
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - P Laforêt
- Nord-Est-Ile de France Neuromuscular Reference Center, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris Myology Institute, Neuromuscular Pathology Reference Center, Groupe Hospitalier Universitaire La Pitié-Salpêtrière, Sorbonne Universités UPMC Univ Paris 06, Paris, France
| | | |
Collapse
|
20
|
Banerji CRS, Zammit PS. Pathomechanisms and biomarkers in facioscapulohumeral muscular dystrophy: roles of DUX4 and PAX7. EMBO Mol Med 2021; 13:e13695. [PMID: 34151531 PMCID: PMC8350899 DOI: 10.15252/emmm.202013695] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 03/27/2021] [Accepted: 03/30/2021] [Indexed: 12/29/2022] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is characterised by progressive skeletal muscle weakness and wasting. FSHD is linked to epigenetic derepression of the subtelomeric D4Z4 macrosatellite at chromosome 4q35. Epigenetic derepression permits the distal-most D4Z4 unit to transcribe DUX4, with transcripts stabilised by splicing to a poly(A) signal on permissive 4qA haplotypes. The pioneer transcription factor DUX4 activates target genes that are proposed to drive FSHD pathology. While this toxic gain-of-function model is a satisfying "bottom-up" genotype-to-phenotype link, DUX4 is rarely detectable in muscle and DUX4 target gene expression is inconsistent in patients. A reliable biomarker for FSHD is suppression of a target gene score of PAX7, a master regulator of myogenesis. However, it is unclear how this "top-down" finding links to genomic changes that characterise FSHD and to DUX4. Here, we explore the roles and interactions of DUX4 and PAX7 in FSHD pathology and how the relationship between these two transcription factors deepens understanding via the immune system and muscle regeneration. Considering how FSHD pathomechanisms are represented by "DUX4opathy" models has implications for developing therapies and current clinical trials.
Collapse
Affiliation(s)
| | - Peter S Zammit
- Randall Centre for Cell and Molecular BiophysicsKing's College LondonLondonUK
| |
Collapse
|
21
|
Nguyen TH, Conotte S, Belayew A, Declèves AE, Legrand A, Tassin A. Hypoxia and Hypoxia-Inducible Factor Signaling in Muscular Dystrophies: Cause and Consequences. Int J Mol Sci 2021; 22:7220. [PMID: 34281273 PMCID: PMC8269128 DOI: 10.3390/ijms22137220] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/28/2021] [Accepted: 06/30/2021] [Indexed: 12/29/2022] Open
Abstract
Muscular dystrophies (MDs) are a group of inherited degenerative muscle disorders characterized by a progressive skeletal muscle wasting. Respiratory impairments and subsequent hypoxemia are encountered in a significant subgroup of patients in almost all MD forms. In response to hypoxic stress, compensatory mechanisms are activated especially through Hypoxia-Inducible Factor 1 α (HIF-1α). In healthy muscle, hypoxia and HIF-1α activation are known to affect oxidative stress balance and metabolism. Recent evidence has also highlighted HIF-1α as a regulator of myogenesis and satellite cell function. However, the impact of HIF-1α pathway modifications in MDs remains to be investigated. Multifactorial pathological mechanisms could lead to HIF-1α activation in patient skeletal muscles. In addition to the genetic defect per se, respiratory failure or blood vessel alterations could modify hypoxia response pathways. Here, we will discuss the current knowledge about the hypoxia response pathway alterations in MDs and address whether such changes could influence MD pathophysiology.
Collapse
Affiliation(s)
- Thuy-Hang Nguyen
- Laboratory of Respiratory Physiology, Pathophysiology and Rehabilitation, Research Institute for Health Sciences and Technology, University of Mons, 7000 Mons, Belgium; (T.-H.N.); (S.C.); (A.B.); (A.L.)
| | - Stephanie Conotte
- Laboratory of Respiratory Physiology, Pathophysiology and Rehabilitation, Research Institute for Health Sciences and Technology, University of Mons, 7000 Mons, Belgium; (T.-H.N.); (S.C.); (A.B.); (A.L.)
| | - Alexandra Belayew
- Laboratory of Respiratory Physiology, Pathophysiology and Rehabilitation, Research Institute for Health Sciences and Technology, University of Mons, 7000 Mons, Belgium; (T.-H.N.); (S.C.); (A.B.); (A.L.)
| | - Anne-Emilie Declèves
- Department of Metabolic and Molecular Biochemistry, Research Institute for Health Sciences and Technology, University of Mons, 7000 Mons, Belgium;
| | - Alexandre Legrand
- Laboratory of Respiratory Physiology, Pathophysiology and Rehabilitation, Research Institute for Health Sciences and Technology, University of Mons, 7000 Mons, Belgium; (T.-H.N.); (S.C.); (A.B.); (A.L.)
| | - Alexandra Tassin
- Laboratory of Respiratory Physiology, Pathophysiology and Rehabilitation, Research Institute for Health Sciences and Technology, University of Mons, 7000 Mons, Belgium; (T.-H.N.); (S.C.); (A.B.); (A.L.)
| |
Collapse
|
22
|
Ohmori S, Sawada Y, Saito-Sasaki N, Sato S, Minokawa Y, Sugino H, Nanamori H, Yamamoto K, Okada E, Nakamura M. A Positive Dermcidin Expression Is an Unfavorable Prognostic Marker for Extramammary Paget's Disease. Diagnostics (Basel) 2021; 11:diagnostics11061086. [PMID: 34198581 PMCID: PMC8231974 DOI: 10.3390/diagnostics11061086] [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: 05/08/2021] [Revised: 06/08/2021] [Accepted: 06/11/2021] [Indexed: 11/16/2022] Open
Abstract
Extramammary Paget’s disease is recognized as an apocrine-origin cutaneous tumor and is localized in the intraepithelial skin lesion. However, its advanced form is intractable, and there is currently no therapeutic option with a satisfactory level of clinical outcome. Therefore, it is of great importance to identify a potential biomarker to estimate tumor advancement in extramammary Paget’s disease. Dermcidin is an antimicrobial peptide derived from the eccrine gland and is identified as a biomarker in various malignancies. To investigate the potential of dermcidin in extramammary Paget’s disease, we investigated dermcidin expression in tumors using the immunostaining technique. Although previous studies have reported that extramammary Paget’s disease has no positive staining against dermcidin, 14 out of 60 patients showed positive staining of dermcidin in our study. To clarify the characteristics of positive dermcidin in extramammary Paget’s disease, we investigated the clinical characteristics of positive dermcidin extramammary Paget’s disease patients. Positive dermcidin patients showed a significantly high frequency of lymph node metastasis. We next investigated the impact of positive dermcidin on overall survival. Univariate analysis identified that positive dermcidin showed a significantly increased hazard ratio in overall survival, suggesting that dermcidin might be a prognostic factor for extramammary Paget’s disease.
Collapse
Affiliation(s)
| | - Yu Sawada
- Correspondence: ; Tel.: +81-093-691-7445
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Redox Homeostasis in Muscular Dystrophies. Cells 2021; 10:cells10061364. [PMID: 34205993 PMCID: PMC8229249 DOI: 10.3390/cells10061364] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 12/15/2022] Open
Abstract
In recent years, growing evidence has suggested a prominent role of oxidative stress in the pathophysiology of several early- and adult-onset muscle disorders, although effective antioxidant treatments are still lacking. Oxidative stress causes cell damage by affecting protein function, membrane structure, lipid metabolism, and DNA integrity, thus interfering with skeletal muscle homeostasis and functionality. Some features related to oxidative stress, such as chronic inflammation, defective regeneration, and mitochondrial damage are shared among most muscular dystrophies, and Nrf2 has been shown to be a central player in antagonizing redox imbalance in several of these disorders. However, the exact mechanisms leading to overproduction of reactive oxygen species and deregulation in the cellular antioxidants system seem to be, to a large extent, disease-specific, and the clarification of these mechanisms in vivo in humans is the cornerstone for the development of targeted antioxidant therapies, which will require testing in appropriately designed clinical trials.
Collapse
|