1
|
Russo C, Surdo S, Valle MS, Malaguarnera L. The Gut Microbiota Involvement in the Panorama of Muscular Dystrophy Pathogenesis. Int J Mol Sci 2024; 25:11310. [PMID: 39457092 PMCID: PMC11508360 DOI: 10.3390/ijms252011310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/16/2024] [Accepted: 10/19/2024] [Indexed: 10/28/2024] Open
Abstract
Muscular dystrophies (MDs) are genetically heterogeneous diseases characterized by primary skeletal muscle atrophy. The collapse of muscle structure and irreversible degeneration of tissues promote the occurrence of comorbidities, including cardiomyopathy and respiratory failure. Mitochondrial dysfunction leads to inflammation, fibrosis, and adipogenic cellular infiltrates that exacerbate the symptomatology of MD patients. Gastrointestinal disorders and metabolic anomalies are common in MD patients and may be determined by the interaction between the intestine and its microbiota. Therefore, the gut-muscle axis is one of the actors involved in the spread of inflammatory signals to all muscles. In this review, we aim to examine in depth how intestinal dysbiosis can modulate the metabolic state, the immune response, and mitochondrial biogenesis in the course and progression of the most investigated MDs such as Duchenne Muscular Dystrophy (DMD) and Myotonic Dystrophy (MD1), to better identify gut microbiota metabolites working as therapeutic adjuvants to improve symptoms of MD.
Collapse
Affiliation(s)
- Cristina Russo
- Section of Pathology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
| | - Sofia Surdo
- Italian Center for the Study of Osteopathy (CSDOI), 95124 Catania, Italy;
| | - Maria Stella Valle
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy;
| | - Lucia Malaguarnera
- Section of Pathology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
| |
Collapse
|
2
|
Finsterer J, Strobl W. Gastrointestinal involvement in neuromuscular disorders. J Gastroenterol Hepatol 2024; 39:1982-1993. [PMID: 38859699 DOI: 10.1111/jgh.16650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/30/2024] [Accepted: 05/25/2024] [Indexed: 06/12/2024]
Abstract
Although not often discussed, many of the neuromuscular disorders (NMDs) affect the gastrointestinal tract (GIT). Depending on the type of NMD, the prevalence of GIT involvement ranges from <5% (e.g. hereditary neuropathies, myofibrillar myopathies) to 100% (e.g. MNGIE, OPMD). Particularly in NMDs with multisystem affection, involvement of the GIT can dominate the clinical presentation or at least make up a significant part of the clinical picture. The most prominent representatives of NMDs with multisystem involvement are the mitochondrial disorders (MIDs) and the myotonic dystrophies. The best known syndromic MIDs with GIT involvement are MNGIE, MELAS, Leigh, and Pearson syndromes. Among neuropathies, GIT involvement is most commonly found in ALS and GBS. GIT involvement may also be a feature of myasthenia. The clinical manifestations of GIT involvement are diverse and can affect the entire GIT, from the teeth to the rectum, including the liver and pancreas. The most well-known clinical manifestations of GIT involvement are dysphagia, nausea, vomiting, reflux, hollow organ dysmotility, hepatopathy, diabetes, diarrhea, constipation, and fecal incontinence. Even if treatment can usually only be symptomatic, the therapeutic options are diverse, are often effective, and can significantly and beneficially influence the course of the underlying NMD.
Collapse
Affiliation(s)
| | - Walter Strobl
- Department of Health Sciences, Medicine and Research, Danube University for Continuing Education Krems and MOTIO, Vienna, Austria
| |
Collapse
|
3
|
Schiava M, Lofra RM, Bourke JP, James MK, Díaz-Manera J, Elseed MA, Michel-Sodhi J, Moat D, Mccallum M, Mayhew A, Ghimenton E, Díaz CFB, Malinova M, Wong K, Richardson M, Tasca G, Grover E, Robinson EJ, Tanner S, Eglon G, Behar L, Eagle M, Turner C, Verdú-Díaz J, Heslop E, Straub V, Bettolo CM, Guglieri M. Disease-associated comorbidities, medication records and anthropometric measures in adults with Duchenne muscular dystrophy. Neuromuscul Disord 2024; 41:8-19. [PMID: 38865917 DOI: 10.1016/j.nmd.2024.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/10/2024] [Accepted: 05/13/2024] [Indexed: 06/14/2024]
Abstract
We investigated the comorbidities, associated factors, and the relationship between anthropometric measures and respiratory function and functional abilities in adults with Duchenne muscular dystrophy (DMD). This was a single-centre cross-sectional study in genetically diagnosed adults with DMD (>16 years old). Univariate and multivariate analyses identified factors associated with dysphagia, constipation, Body Mass Index (BMI), and weight. Regression analysis explored associations between BMI, weight, and respiratory/motor abilities. We included 112 individuals (23.4 ± 5.2 years old), glucocorticoid-treated 66.1 %. The comorbidities frequency was 61.6 % scoliosis (61.0 % of them had spinal surgery), 36.6 % dysphagia, 36.6 % constipation, and 27.8 % urinary conditions. The use of glucocorticoids delayed the time to spinal surgery. The univariate analysis revealed associations between dysphagia and constipation with age, lack of glucocorticoid treatment, and lower respiratory and motor function. In the multivariate analysis, impaired cough ability remained as the factor consistently linked to both conditions. Constipation associated with lower BMI and weight. BMI and weight positively correlated with respiratory parameters, but they did not associate with functional abilities. Glucocorticoids reduce the frequency of comorbidities in adults with DMD. The ability to cough can help identifying dysphagia and constipation. Lower BMI and weight in individuals with DMD with compromised respiratory function may suggest a higher calories requirement.
Collapse
Affiliation(s)
- Marianela Schiava
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Robert Muni Lofra
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - John P Bourke
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Meredith K James
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Jordi Díaz-Manera
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Maha A Elseed
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Jassi Michel-Sodhi
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Dionne Moat
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Michelle Mccallum
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Anna Mayhew
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Elisabetta Ghimenton
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Carla Florencia Bolaño Díaz
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Monika Malinova
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Karen Wong
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Mark Richardson
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Giorgio Tasca
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Emma Grover
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Emma-Jayne Robinson
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Stephanie Tanner
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Gail Eglon
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Laura Behar
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | | | - Catherine Turner
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - José Verdú-Díaz
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Emma Heslop
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Volker Straub
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Chiara Marini Bettolo
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Michela Guglieri
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK.
| |
Collapse
|
4
|
Subhan F, Zizzo MG, Serio R. Motor dysfunction of the gut in Duchenne muscular dystrophy: A review. Neurogastroenterol Motil 2024; 36:e14804. [PMID: 38651673 DOI: 10.1111/nmo.14804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 04/06/2024] [Accepted: 04/09/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND Duchenne's muscular dystrophy (DMD) is a severe type of hereditary, neuromuscular disorder caused by a mutation in the dystrophin gene resulting in the absence or production of truncated dystrophin protein. Conventionally, clinical descriptions of the disorder focus principally on striated muscle defects; however, DMD manifestations involving gastrointestinal (GI) smooth muscle have been reported, even if not rigorously studied. PURPOSE The objective of the present review is to offer a comprehensive perspective on the existing knowledge concerning GI manifestations in DMD, focusing the attention on evidence in DMD patients and mdx mice. This includes an assessment of symptomatology, etiological pathways, and potential corrective approaches. This paper could provide helpful information about DMD gastrointestinal implications that could serve as a valuable orientation for prospective research endeavors in this field. This manuscript emphasizes the effectiveness of mdx mice, a DMD animal model, in unraveling mechanistic insights and exploring the pathological alterations in the GI tract. The gastrointestinal consequences evident in patients with DMD and the mdx mice models are a significant area of focus for researchers. The exploration of this area in depth could facilitate the development of more efficient therapeutic approaches and improve the well-being of individuals impacted by the condition.
Collapse
Affiliation(s)
- Fazal Subhan
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, Palermo, Italy
| | - Maria Grazia Zizzo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, Palermo, Italy
- ATeN (Advanced Technologies Network) Center, Viale delle Scienze, University of Palermo, Palermo, Italy
| | - Rosa Serio
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, Palermo, Italy
| |
Collapse
|
5
|
Konieczny P. Systemic Treatment of Body-Wide Duchenne Muscular Dystrophy Symptoms. Clin Pharmacol Ther 2024. [PMID: 38965715 DOI: 10.1002/cpt.3363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 06/09/2024] [Indexed: 07/06/2024]
Abstract
Duchenne muscular dystrophy (DMD) is a fatal X-linked disease that leads to premature death due to the loss of dystrophin. Current strategies predominantly focus on the therapeutic treatment of affected skeletal muscle tissue. However, certain results point to the fact that with successful treatment of skeletal muscle, DMD-exposed latent phenotypes in tissues, such as cardiac and smooth muscle, might lead to adverse effects and even death. Likewise, it is now clear that the absence of dystrophin affects the function of the nervous system, and that this phenotype is more pronounced when shorter dystrophins are absent, in addition to the full-length dystrophin that is present predominantly in the muscle. Here, I focus on the systemic aspects of DMD, highlighting the ubiquitous expression of the dystrophin gene in human tissues. Furthermore, I describe therapeutic strategies that have been tested in the clinic and point to unresolved questions regarding the function of distinct dystrophin isoforms, and the possibility of current therapeutic strategies to tackle phenotypes that relate to their absence.
Collapse
Affiliation(s)
- Patryk Konieczny
- Institute of Human Biology and Evolution, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| |
Collapse
|
6
|
Blokhuis AM, Tytgat K, Groothuis JT, Houwen-van Opstal S. Severe gastrointestinal problems in Duchenne muscular dystrophy: A case series. Neuromuscul Disord 2024; 40:31-37. [PMID: 38823288 DOI: 10.1016/j.nmd.2024.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 05/10/2024] [Accepted: 05/10/2024] [Indexed: 06/03/2024]
Abstract
Due to improved supportive care, survival of patients with Duchenne muscular dystrophy (DMD) has increased significantly. Consequently, new challenges emerge in adult patients with DMD. In clinical practice we increasingly see patients with serious, even life-threatening, gastrointestinal (GI) problems in advanced disease stages. Little is known about the longitudinal course of GI problems and the appropriate management. We present a case-series of six adult patients with DMD with (recurrent) GI problems that required hospital admission. The most prevalent reported serious GI symptoms were gastrointestinal pseudo-obstruction, (sub)ileus and gastric dilatation. Besides, an overview is presented of the therapeutic options for GI problems in DMD. The current study provides insight in possible treatment options, however, there is a clear need for more research and an integral guideline on treatment of GI problems in adult patients with DMD in order to reduce associated morbidity and mortality.
Collapse
Affiliation(s)
- A M Blokhuis
- Department of Rehabilitation, Sint Maartenskliniek, Nijmegen, the Netherlands
| | - Kmaj Tytgat
- Department of Gastroenterology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - J T Groothuis
- Department of Rehabilitation, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands; Radboudumc Center of Expertise for neuromuscular disorders (Radboud-NMD), Netherlands Neuromuscular Center (NL-NMD) and the European Reference Network for rare neuromuscular diseases (EURO-NMD)
| | - Sls Houwen-van Opstal
- Department of Rehabilitation, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Amalia Children's Hospital, Postbus 9101, Huispostnr 898 HB, Nijmegen 6500, the Netherlands; Radboudumc Center of Expertise for neuromuscular disorders (Radboud-NMD), Netherlands Neuromuscular Center (NL-NMD) and the European Reference Network for rare neuromuscular diseases (EURO-NMD).
| |
Collapse
|
7
|
Dubinin MV, Mikheeva IB, Stepanova AE, Pavlova EK, Gazheeva TP, Belosludtsev KN. Glucocorticoid Deflazacort Normalizes the Ultrastructure of Skeletal Muscles and the State of the Colon Microbiota in Dystrophin-Deficient Mice. Bull Exp Biol Med 2024; 177:333-338. [PMID: 39126545 DOI: 10.1007/s10517-024-06184-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Indexed: 08/12/2024]
Abstract
We studied the effect of enteral administration of the glucocorticoid deflazacort (DFC, 1.2 mg/kg per day, 28 days) on the state of skeletal muscles and tissue ultrastructure, as well as the composition of the colon microbiota in dystrophin-deficient mdx mice. DFC has been shown to reduce the intensity of degeneration/regeneration cycles in muscle fibers of mdx mice. This effect of DFC was accompanied by normalization of the size of sarcomeres of skeletal muscles of mdx mice, improvement of the ultrastructure of the subsarcolemmal population of mitochondria, and an increase in the number of organelles, as well as normalization of the number of contact interactions between the sarcoplasmic reticulum and mitochondria. In addition, DFC had a corrective effect on the colon microbiota of mdx mice, which manifested in an increase in the number of the Bifidobacterium genus microorganisms and a decrease in the level of E. coli with reduced enzymatic activity.
Collapse
Affiliation(s)
- M V Dubinin
- Mari State University, Yoshkar-Ola, Mari El Republic, Russia.
| | - I B Mikheeva
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow region, Russia
| | - A E Stepanova
- Mari State University, Yoshkar-Ola, Mari El Republic, Russia
| | - E K Pavlova
- Mari State University, Yoshkar-Ola, Mari El Republic, Russia
| | - T P Gazheeva
- Mari State University, Yoshkar-Ola, Mari El Republic, Russia
| | - K N Belosludtsev
- Mari State University, Yoshkar-Ola, Mari El Republic, Russia
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow region, Russia
| |
Collapse
|
8
|
Jollet M, Mariadassou M, Rué O, Pessemesse L, Ollendorff V, Ramdani S, Vernus B, Bonnieu A, Bertrand-Gaday C, Goustard B, Koechlin-Ramonatxo C. Insight into the Role of Gut Microbiota in Duchenne Muscular Dystrophy: An Age-Related Study in Mdx Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:264-279. [PMID: 37981219 DOI: 10.1016/j.ajpath.2023.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 10/06/2023] [Accepted: 10/31/2023] [Indexed: 11/21/2023]
Abstract
Dystrophin deficiency alters the sarcolemma structure, leading to muscle dystrophy, muscle disuse, and ultimately death. Beyond limb muscle deficits, patients with Duchenne muscular dystrophy have numerous transit disorders. Many studies have highlighted the strong relationship between gut microbiota and skeletal muscle. The aims of this study were: i) to characterize the gut microbiota composition over time up to 1 year in dystrophin-deficient mdx mice, and ii) to analyze the intestine structure and function and expression of genes linked to bacterial-derived metabolites in ileum, blood, and skeletal muscles to study interorgan interactions. Mdx mice displayed a significant reduction in the overall number of different operational taxonomic units and their abundance (α-diversity). Mdx genotype predicted 20% of β-diversity divergence, with a large taxonomic modification of Actinobacteria, Proteobacteria, Tenericutes, and Deferribacteres phyla and the included genera. Interestingly, mdx intestinal motility and gene expressions of tight junction and Ffar2 receptor were down-regulated in the ileum. Concomitantly, circulating inflammatory markers related to gut microbiota (tumor necrosis factor, IL-6, monocyte chemoattractant protein-1) and muscle inflammation Tlr4/Myd88 pathway (Toll-like receptor 4, which recognizes pathogen-associated molecular patterns) were up-regulated. Finally, in mdx mice, adiponectin was reduced in blood and its receptor modulated in muscles. This study highlights a specific gut microbiota composition and highlights interorgan interactions in mdx physiopathology with gut microbiota as the potential central metabolic organ.
Collapse
Affiliation(s)
- Maxence Jollet
- DMEM, Université de Montpellier, INRAE, Montpellier, France.
| | - Mahendra Mariadassou
- Université Paris-Saclay, INRAE, BioinfOmics, MIGALE Bioinformatics Facility, Jouy-en-Josas, France; Université Paris-Saclay, INRAE, MaIAGE, Jouy-en-Josas, France
| | - Olivier Rué
- Université Paris-Saclay, INRAE, BioinfOmics, MIGALE Bioinformatics Facility, Jouy-en-Josas, France; Université Paris-Saclay, INRAE, MaIAGE, Jouy-en-Josas, France
| | | | | | | | - Barbara Vernus
- DMEM, Université de Montpellier, INRAE, Montpellier, France
| | - Anne Bonnieu
- DMEM, Université de Montpellier, INRAE, Montpellier, France
| | | | | | | |
Collapse
|
9
|
Dowling P, Trollet C, Negroni E, Swandulla D, Ohlendieck K. How Can Proteomics Help to Elucidate the Pathophysiological Crosstalk in Muscular Dystrophy and Associated Multi-System Dysfunction? Proteomes 2024; 12:4. [PMID: 38250815 PMCID: PMC10801633 DOI: 10.3390/proteomes12010004] [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: 12/05/2023] [Revised: 01/09/2024] [Accepted: 01/12/2024] [Indexed: 01/23/2024] Open
Abstract
This perspective article is concerned with the question of how proteomics, which is a core technique of systems biology that is deeply embedded in the multi-omics field of modern bioresearch, can help us better understand the molecular pathogenesis of complex diseases. As an illustrative example of a monogenetic disorder that primarily affects the neuromuscular system but is characterized by a plethora of multi-system pathophysiological alterations, the muscle-wasting disease Duchenne muscular dystrophy was examined. Recent achievements in the field of dystrophinopathy research are described with special reference to the proteome-wide complexity of neuromuscular changes and body-wide alterations/adaptations. Based on a description of the current applications of top-down versus bottom-up proteomic approaches and their technical challenges, future systems biological approaches are outlined. The envisaged holistic and integromic bioanalysis would encompass the integration of diverse omics-type studies including inter- and intra-proteomics as the core disciplines for systematic protein evaluations, with sophisticated biomolecular analyses, including physiology, molecular biology, biochemistry and histochemistry. Integrated proteomic findings promise to be instrumental in improving our detailed knowledge of pathogenic mechanisms and multi-system dysfunction, widening the available biomarker signature of dystrophinopathy for improved diagnostic/prognostic procedures, and advancing the identification of novel therapeutic targets to treat Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Paul Dowling
- Department of Biology, Maynooth University, National University of Ireland, W23 F2H6 Maynooth, Co. Kildare, Ireland;
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, W23 F2H6 Maynooth, Co. Kildare, Ireland
| | - Capucine Trollet
- Center for Research in Myology U974, Sorbonne Université, INSERM, Myology Institute, 75013 Paris, France; (C.T.); (E.N.)
| | - Elisa Negroni
- Center for Research in Myology U974, Sorbonne Université, INSERM, Myology Institute, 75013 Paris, France; (C.T.); (E.N.)
| | - Dieter Swandulla
- Institute of Physiology, Faculty of Medicine, University of Bonn, D53115 Bonn, Germany;
| | - Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, W23 F2H6 Maynooth, Co. Kildare, Ireland;
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, W23 F2H6 Maynooth, Co. Kildare, Ireland
| |
Collapse
|
10
|
Gadaleta G, Urbano G, Brusa C, D'Alessandro R, Rolle E, Cavallina I, Mattei A, Ribolla F, Raineri C, Pidello S, Vercelli L, Ricci FS, Mongini TE. Adults living with Duchenne muscular dystrophy: old and new challenges in a cohort of 19 patients in their third to fifth decade. Eur J Neurol 2024; 31:e16060. [PMID: 37724986 PMCID: PMC11235709 DOI: 10.1111/ene.16060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 08/02/2023] [Accepted: 08/30/2023] [Indexed: 09/21/2023]
Abstract
BACKGROUND AND PURPOSE Advances in multidisciplinary care are extending overall survival in Duchenne muscular dystrophy (DMD) patients. Our research objective was to delineate the clinical characteristics of this particular cohort and identify novel challenges associated with the disease. METHODS Nineteen individuals aged 25-48 years (median 34 years) with a confirmed diagnosis of out-of-frame DMD gene mutation were selected. RESULTS All patients were mechanically ventilated (5/19 via tracheostomy), with different patterns of cardiomyopathy. Swallowing and nutritional issues were frequent (median body mass index 18.95), with six cases requiring artificial enteral feeding (median age at start 29 years), as well as bone density alterations (11/19, 58%). Only 2/19 had been on long-term prednisone therapy. Issues requiring at-home/hospital assistance were respiratory infections (15/19, 79%), gastroenterological symptoms (9/19, 47%, including toxic megacolon and rectal perforation after repeated enemas), metabolic acidosis (2/19, 11%) and recurrent ischaemic strokes (1/19, 5%). From a social perspective, augmented-alternative communication devices were necessary for 7/19 (37%), with most of the patients being assisted at home and 2/19 institutionalized. Eight/19 (42%) patients experienced psychiatric symptoms (median age at presentation 16 years) and 9/19 (47%) chronic pain (median age at onset 23 years), in both cases treated with psychoactive/analgesic drugs without major adverse events. The patients' subjective perception of physical health resulted in unfavourable scores, whilst the subjective assessment of mental health unexpectedly showed more positive values compared to other chronic neurological conditions. CONCLUSIONS The analysis of adults living with DMD reveals several new health-related issues, such as the management of emergencies and safety of pharmacological treatments for psychiatric symptoms, chronic pain management, as well as an increasing caregivers burden.
Collapse
Affiliation(s)
- Giulio Gadaleta
- Neuromuscular Unit, Department of Neurosciences ‘Rita Levi Montalcini’University of TurinTurinItaly
| | - Guido Urbano
- Neuromuscular Unit, Department of Neurosciences ‘Rita Levi Montalcini’University of TurinTurinItaly
| | - Chiara Brusa
- Neuromuscular Unit, Department of Neurosciences ‘Rita Levi Montalcini’University of TurinTurinItaly
- Section of Child and Adolescent Neuropsychiatry, Department of Public Health and Paediatric SciencesUniversity of TurinTurinItaly
- Dubowitz Neuromuscular Centre, University College LondonGreat Ormond Street Institute of Child HealthLondonUK
| | - Rossella D'Alessandro
- Section of Child and Adolescent Neuropsychiatry, Department of Public Health and Paediatric SciencesUniversity of TurinTurinItaly
| | - Enrica Rolle
- Neuromuscular Unit, Department of Neurosciences ‘Rita Levi Montalcini’University of TurinTurinItaly
- Section of Child and Adolescent Neuropsychiatry, Department of Public Health and Paediatric SciencesUniversity of TurinTurinItaly
| | - Ilaria Cavallina
- Neuromuscular Unit, Department of Neurosciences ‘Rita Levi Montalcini’University of TurinTurinItaly
- Section of Child and Adolescent Neuropsychiatry, Department of Public Health and Paediatric SciencesUniversity of TurinTurinItaly
| | | | - Fulvia Ribolla
- Pneumology Unit, Department of Cardiovascular and Thoracic MedicineA.O.U. Città della Salute e della Scienza di TorinoTurinItaly
| | - Claudia Raineri
- Division of Cardiology, Department of Cardiovascular and Thoracic MedicineA.O.U. Città della Salute e della Scienza di TorinoTurinItaly
| | - Stefano Pidello
- Division of Cardiology, Department of Cardiovascular and Thoracic MedicineA.O.U. Città della Salute e della Scienza di TorinoTurinItaly
| | - Liliana Vercelli
- Neuromuscular Unit, Department of Neurosciences ‘Rita Levi Montalcini’University of TurinTurinItaly
| | - Federica S. Ricci
- Section of Child and Adolescent Neuropsychiatry, Department of Public Health and Paediatric SciencesUniversity of TurinTurinItaly
| | - Tiziana E. Mongini
- Neuromuscular Unit, Department of Neurosciences ‘Rita Levi Montalcini’University of TurinTurinItaly
| |
Collapse
|
11
|
Nart L, Desikan M, Emmanuel A, Quinlivan R. Life-threatening bowel complications in adults with Duchenne muscular dystrophy: a case series. Neuromuscul Disord 2023; 33:873-876. [PMID: 37919206 DOI: 10.1016/j.nmd.2023.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 10/02/2023] [Accepted: 10/12/2023] [Indexed: 11/04/2023]
Abstract
Duchenne muscular dystrophy is a progressive muscle wasting disease caused by pathogenic variants in DMD. Gastrointestinal involvement is increasingly recognised in older patients and can manifest as life-threatening bowel dysmotility. We describe a series of adults with Duchenne muscular dystrophy who developed either severe colonic pseudo-obstruction or sigmoid volvulus requiring urgent assessment and intervention. The presentations varied in their clinical picture and outcomes, but together highlight the complexity of managing gastrointestinal complications in this cohort of patients. Key considerations include pre-existing cardiorespiratory compromise and the increased risk associated with surgery and general anaesthesia. We also outline a role for home parenteral nutrition in the long-term management of associated bowel dysmotility.
Collapse
Affiliation(s)
- Luca Nart
- Neuromuscular Complex Care Centre, National Hospital for Neurology and Neurosurgery, Queen Square, London, United Kingdom.
| | - Mahalekshmi Desikan
- Neuromuscular Complex Care Centre, National Hospital for Neurology and Neurosurgery, Queen Square, London, United Kingdom
| | - Anton Emmanuel
- Gastrointestinal Physiology Unit, University College London Hospitals, London, United Kingdom
| | - Ros Quinlivan
- Neuromuscular Complex Care Centre, National Hospital for Neurology and Neurosurgery, Queen Square, London, United Kingdom; Gastrointestinal Physiology Unit, University College London Hospitals, London, United Kingdom; MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, London, United Kingdom
| |
Collapse
|
12
|
Ramicone I, Whitford B, Mosley L, Thienprayoon R. Polysymptomatology and Polypharmacy at End of Life in Patients with Duchenne Muscular Dystrophy. J Pediatr 2023; 261:113598. [PMID: 37402439 DOI: 10.1016/j.jpeds.2023.113598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 07/06/2023]
Abstract
OBJECTIVE To characterize symptom frequency and symptom-directed treatment approaches in patients who died with advanced Duchenne muscular dystrophy (DMD). STUDY DESIGN This was a retrospective cohort study of patients in a multidisciplinary DMD program who died between January 1, 2013, and June 30, 2021. Inclusion criteria were patients who died with advanced DMD in the time period studied; exclusion criteria were low exposure to palliative care (<2 encounters). Demographic, symptom, and end-of-life data, as well as medications used for symptom management, were abstracted from the electronic medical record. RESULTS In total, 15 patients were eligible for analysis. The median age of death was 23 years (range 15-30 years). One (6.7%) experienced a full code at death, 8 (53.3%) had do-not-resuscitate orders, and 4 (26.7%) had limited do-not-resuscitate orders. Mean palliative care exposure was 1280 days. All 15 (100%) had pain and dyspnea; 14 (93.3%) anorexia, constipation, and sleep difficulty; 13 (86.7%) wounds; and 12 (80%) anxiety and nausea/vomiting. Multiple medications and drug classes were used to target symptoms. CONCLUSIONS We found significant polysymptomatology and polypharmacy in patients who died with advanced DMD. Clinicians who care for patients with advanced DMD should clarify goals of care and document advance care planning. Given the complexity of multisystem disease progression, palliative care should provide subspecialty pain management and assist with psychosocial burdens.
Collapse
Affiliation(s)
- Isabella Ramicone
- Division of Palliative Care, Department of Anesthesia, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Brittney Whitford
- Division of Palliative Care, Department of Anesthesia, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Luke Mosley
- Division of Palliative Care, Department of Anesthesia, University of Cincinnati College of Medicine, Cincinnati, OH; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Rachel Thienprayoon
- Division of Palliative Care, Department of Anesthesia, University of Cincinnati College of Medicine, Cincinnati, OH; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH.
| |
Collapse
|
13
|
Lechner A, Herzig JJ, Kientsch JG, Kohler M, Bloch KE, Ulrich S, Schwarz EI. Cardiomyopathy as cause of death in Duchenne muscular dystrophy: a longitudinal observational study. ERJ Open Res 2023; 9:00176-2023. [PMID: 37727676 PMCID: PMC10505954 DOI: 10.1183/23120541.00176-2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 07/06/2023] [Indexed: 09/21/2023] Open
Abstract
Background Cardiomyopathy has become an important life-limiting factor since survival in Duchenne muscular dystrophy (DMD) has greatly increased with long-term ventilation and cough assistance. The aim of this study was to investigate the association between impaired left ventricular ejection fraction (LVEF) and survival. Methods In a >20-year observational study in patients with DMD (age ≥16 years) with at least three echocardiograms, the association between LVEF and survival and time to cardiac or non-cardiac death was investigated using Kaplan-Meier survival analysis and Cox regression (for LVEF). Results In 67 DMD patients (430 echocardiograms), the decrease in LVEF over a mean±sd follow-up period of 9.1±5.1 years was -10.0±13.9% absolute, but LVEF progression varied widely. 84% were receiving an angiotensin-converting enzyme inhibitor and 54% a β-blocker at last follow-up with an LVEF of 37.5±12.4% at that time-point. Median (interquartile range) survival was 33 (25-40) years. 28 out of 67 (42%) of the cohort had died and LVEF was a significant negative predictor of survival (hazard ratio 0.95 (95% CI 0.91-0.99); p<0.007). Those who died of cardiac death (53% of known causes of death) had significantly lower LVEF at the time of death (LVEF -11.0% (95% CI -21.1- -0.9%); p=0.035) compared with non-cardiac death and tended to die at a younger age. Conclusions Cardiomyopathy with systolic heart failure is the leading cause of death and lower LVEF is an independent predictor of mortality at younger ages in patients with DMD. Patients with DMD appear to be undertreated with respect to heart failure drug therapy.
Collapse
Affiliation(s)
- Annabel Lechner
- Department of Pulmonology and Sleep Disorders Centre, University Hospital of Zurich (USZ), Zurich, Switzerland
| | - Joël J. Herzig
- Department of Pulmonology and Sleep Disorders Centre, University Hospital of Zurich (USZ), Zurich, Switzerland
| | - Jacqueline G. Kientsch
- Department of Pulmonology and Sleep Disorders Centre, University Hospital of Zurich (USZ), Zurich, Switzerland
| | - Malcolm Kohler
- Department of Pulmonology and Sleep Disorders Centre, University Hospital of Zurich (USZ), Zurich, Switzerland
- Competence Centre Sleep and Health, University of Zurich (UZH), Zurich, Switzerland
| | - Konrad E. Bloch
- Department of Pulmonology and Sleep Disorders Centre, University Hospital of Zurich (USZ), Zurich, Switzerland
- Competence Centre Sleep and Health, University of Zurich (UZH), Zurich, Switzerland
| | - Silvia Ulrich
- Department of Pulmonology and Sleep Disorders Centre, University Hospital of Zurich (USZ), Zurich, Switzerland
| | - Esther I. Schwarz
- Department of Pulmonology and Sleep Disorders Centre, University Hospital of Zurich (USZ), Zurich, Switzerland
- Competence Centre Sleep and Health, University of Zurich (UZH), Zurich, Switzerland
| |
Collapse
|
14
|
Marullo AL, O'Halloran KD. Microbes, metabolites and muscle: Is the gut-muscle axis a plausible therapeutic target in Duchenne muscular dystrophy? Exp Physiol 2023; 108:1132-1143. [PMID: 37269541 PMCID: PMC10988500 DOI: 10.1113/ep091063] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 05/17/2023] [Indexed: 06/05/2023]
Abstract
NEW FINDINGS What is the topic of this review? The contribution of gut microbial signalling to skeletal muscle maintenance and development and identification of potential therapeutic targets in progressive muscle degenerative diseases such as Duchenne muscular dystrophy. What advances does it highlight? Gut microbe-derived metabolites are multifaceted signalling molecules key to muscle function, modifying pathways contributing to skeletal muscle wasting, making them a plausible target for adjunctive therapy in muscular dystrophy. ABSTRACT Skeletal muscle is the largest metabolic organ making up ∼50% of body mass. Because skeletal muscle has both metabolic and endocrine properties, it can manipulate the microbial populations within the gut. In return, microbes exert considerable influence on skeletal muscle via numerous signalling pathways. Gut bacteria produce metabolites (i.e., short chain fatty acids, secondary bile acids and neurotransmitter substrates) that act as fuel sources and modulators of inflammation, influencing host muscle development, growth and maintenance. The reciprocal interactions between microbes, metabolites and muscle establish a bidirectional gut-muscle axis. The muscular dystrophies constitute a broad range of disorders with varying disabilities. In the profoundly debilitating monogenic disorder Duchenne muscular dystrophy (DMD), skeletal muscle undergoes a reduction in muscle regenerative capacity leading to progressive muscle wasting, resulting in fibrotic remodelling and adipose infiltration. The loss of respiratory muscle in DMD culminates in respiratory insufficiency and eventually premature death. The pathways contributing to aberrant muscle remodelling are potentially modulated by gut microbial metabolites, thus making them plausible targets for pre- and probiotic supplementation. Prednisone, the gold standard therapy for DMD, drives gut dysbiosis, inducing a pro-inflammatory phenotype and leaky gut barrier contributing to several of the well-known side effects associated with chronic glucocorticoid treatment. Several studies have observed that gut microbial supplementation or transplantation exerts positive effects on muscle, including mitigating the side effects of prednisone. There is growing evidence in support of the potential for an adjunctive microbiota-directed regimen designed to optimise gut-muscle axis signalling, which could alleviate muscle wasting in DMD.
Collapse
Affiliation(s)
- Anthony L. Marullo
- Department of Physiology, School of Medicine, College of Medicine and HealthUniversity College CorkCorkIreland
| | - Ken D. O'Halloran
- Department of Physiology, School of Medicine, College of Medicine and HealthUniversity College CorkCorkIreland
| |
Collapse
|
15
|
Lupu M, Ioghen M, Perjoc RȘ, Scarlat AM, Vladâcenco OA, Roza E, Epure DAM, Teleanu RI, Severin EM. The Importance of Implementing a Transition Strategy for Patients with Muscular Dystrophy: From Child to Adult-Insights from a Tertiary Centre for Rare Neurological Diseases. CHILDREN (BASEL, SWITZERLAND) 2023; 10:959. [PMID: 37371191 DOI: 10.3390/children10060959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/16/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023]
Abstract
Progress in the field of muscular dystrophy (MD) using a multidisciplinary approach based on international standards of care has led to a significant increase in the life expectancy of patients. The challenge of transitioning from pediatric to adult healthcare has been acknowledged for over a decade, yet it continues to be a last-minute concern. Currently, there is no established consensus on how to evaluate the effectiveness of the transition process. Our study aimed to identify how well patients are prepared for the transition and to determine their needs. We conducted a descriptive, cross-sectional study on 15 patients aged 14 to 21 years. The patients completed a sociodemographic and a Transition Readiness Assessment Questionnaire (TRAQ). We also analyzed the comorbidities of these patients. Our study revealed that only 46.7% of the patients had engaged in a conversation with a medical professional, namely, a child neurologist, about transitioning. A total of 60% of the participants expressed having confidence in their self-care ability. However, the median TRAQ score of 3.6 shows that these patients overestimate themselves. We emphasize the necessity for a slow, personalized transition led by a multidisciplinary team to ensure the continuity of state-of-the-art care from pediatric to adult healthcare services and the achievement of the highest possible quality of life for these patients.
Collapse
Affiliation(s)
- Maria Lupu
- Clinical Neurosciences Department, Peadiatric Neurology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Mihaela Ioghen
- Clinical Neurosciences Department, Peadiatric Neurology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Radu-Ștefan Perjoc
- Clinical Neurosciences Department, Peadiatric Neurology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Andra-Maria Scarlat
- Clinical Neurosciences Department, Peadiatric Neurology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Oana Aurelia Vladâcenco
- Clinical Neurosciences Department, Peadiatric Neurology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Paediatric Neurology, Dr Victor Gomoiu Children's Hospital, 022102 Bucharest, Romania
| | - Eugenia Roza
- Clinical Neurosciences Department, Peadiatric Neurology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Paediatric Neurology, Dr Victor Gomoiu Children's Hospital, 022102 Bucharest, Romania
| | - Diana Ana-Maria Epure
- Department of Paediatric Neurology, Dr Victor Gomoiu Children's Hospital, 022102 Bucharest, Romania
| | - Raluca Ioana Teleanu
- Clinical Neurosciences Department, Peadiatric Neurology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Paediatric Neurology, Dr Victor Gomoiu Children's Hospital, 022102 Bucharest, Romania
| | - Emilia Maria Severin
- Clinical Neurosciences Department, Peadiatric Neurology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
16
|
Crescimanno G, Greco F, Bertini M, Maltese G, Marrone O. Age Related Burden of Swallowing in Adult Patients Affected by Duchenne Muscular Dystrophy. J Neuromuscul Dis 2023; 10:955-962. [PMID: 37212070 PMCID: PMC10578249 DOI: 10.3233/jnd-230055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/27/2023] [Indexed: 05/23/2023]
Abstract
BACKGROUND In Duchenne muscular dystrophy (DMD), dysphagia is a common but often overlooked symptom, which may affect quality of life (QoL). Its possible causes are progressive deterioration of muscle groups involved in swallowing function (oropharyngeal, inspiratory muscles) or impairment of autonomic function. OBJECTIVES In adult patients with DMD, we aimed to identify predictors of swallowing-related QoL and to compare swallowing-related QoL at different ages. METHODS Forty-eight patients aged 30.0±6.6 years were enrolled. Questionnaires were administered: the Swallowing Quality of Life questionnaire (SWAL-QOL) for swallowing-related QoL assessment, and the Compass 31 for autonomic symptoms assessment. The Brooke Upper Extremity Scale was used for upper limbs muscular function assessment. Respiratory and muscle function tests were performed, including spirometry, arterial blood gases, polysomnography, maximal inspiratory pressure (MIP), maximal expiratory pressure and sniff nasal inspiratory pressure. RESULTS An abnormal composite SWAL-QOL score (≤86) was found in 33 patients. Autonomic symptoms were mild, while a severe impairment was shown by the Brooke Upper Extremity Scale. Spirometry and muscle strength tests demonstrated severe alterations, while diurnal and nocturnal blood gases were normal, due to effective use of noninvasive ventilation. Independent predictors of the composite SWAL-QOL score were age, MIP and Compass 31. A MIP < 22 had an accuracy of 92% in predicting altered swallowing-related QoL. The composite SWAL-QOL score was worse in subjects > 30 years old than in younger patients (64.5±19.2 vs 76.6±16.3, p < 0.02), due to worse scores in items pertinent to mental and social functioning; scores in domains pertinent to the physical function were similar in both groups. CONCLUSIONS In adult DMD, swallowing-related QoL, which is altered in most patients, can be predicted by age, inspiratory muscles strength and autonomic dysfunction symptoms. While swallowing function is already altered in young patients, swallowing-related QoL can progressively worsen with advancing age due to psychological and social factors.
Collapse
Affiliation(s)
- Grazia Crescimanno
- Istituto per la Ricerca e l’Innovazione Biomedica (IRIB), Consiglio Nazionale delle Ricerche (CNR), Palermo, Italy
| | - Francesca Greco
- Unione Italiana contro la Distrofia Muscolare (UILDM), Palermo, Italy
| | - Manuela Bertini
- U.O.C. di Lungodegenza, Ospedali Riuniti Villa Sofia-Cervello, Palermo, Italy
| | - Giacomo Maltese
- Unione Italiana contro la Distrofia Muscolare (UILDM), Palermo, Italy
| | - Oreste Marrone
- Istituto per la Ricerca e l’Innovazione Biomedica (IRIB), Consiglio Nazionale delle Ricerche (CNR), Palermo, Italy
| |
Collapse
|
17
|
Corsello A, Scatigno L, Govoni A, Zuccotti G, Gottrand F, Romano C, Verduci E. Gut dysmotility in children with neurological impairment: the nutritional management. Front Neurol 2023; 14:1200101. [PMID: 37213895 PMCID: PMC10196023 DOI: 10.3389/fneur.2023.1200101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 04/20/2023] [Indexed: 05/23/2023] Open
Abstract
Intestinal motility disorders represent a frequent problem in children with neurological impairment. These conditions are characterized by abnormal movements of the gut, which can result in symptoms such as constipation, diarrhea, reflux, and vomiting. The underlying mechanisms leading to dysmotility are various, and the clinical manifestations are often nonspecific. Nutritional management is an important aspect of care for children with gut dysmotility, as it can help to improve their quality of life. Oral feeding, when safe and in the absence of risk of ingestion or severe dysphagia, should always be encouraged. When oral nutrition is insufficient or potentially harmful, it is necessary to switch to an enteral by tube or parenteral nutrition before the onset of malnutrition. In most cases, children with severe gut dysmotility may require feeding via a permanent gastrostomy tube to ensure adequate nutrition and hydration. Drugs may be necessary to help manage gut dysmotility, such as laxatives, anticholinergics and prokinetic agents. Nutritional management of patients with neurological impairment often requires an individualized care plan to optimize growth and nutrition and to improve overall health outcomes. This review tries to sum up most significant neurogenetic and neurometabolic disorders associated with gut dysmotility that may require a specific multidisciplinary care, identifying a proposal of nutritional and medical management.
Collapse
Affiliation(s)
- Antonio Corsello
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, Milan, Italy
| | - Lorenzo Scatigno
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, Milan, Italy
| | - Annalisa Govoni
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, Milan, Italy
| | - Gianvincenzo Zuccotti
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, Milan, Italy
- Department of Biomedical and Clinical Sciences L. Sacco, University of Milan, Milan, Italy
| | - Frédéric Gottrand
- Department of Pediatric Gastroenterology, Hepatology, and Nutrition, CHU Lille, University of Lille, Lille, France
| | - Claudio Romano
- Pediatric Gastroenterology and Cystic Fibrosis Unit, Department of Human Pathology in Adulthood and Childhood "G. Barresi", University of Messina, Messina, Italy
| | - Elvira Verduci
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, University of Milan, Milan, Italy
- Department of Health Science, University of Milan, Milan, Italy
- *Correspondence: Elvira Verduci,
| |
Collapse
|
18
|
Watson-Fargie T, Raeside D, Davidson S, McCartney R, Clarke A, Farrugia ME. Morbidity and Mortality Associated with Gastrointestinal Dysfunction in Neuromuscular Disease: A Single-Centre Case Series. Neuromuscul Disord 2022; 32:578-581. [DOI: 10.1016/j.nmd.2022.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 05/07/2022] [Accepted: 05/11/2022] [Indexed: 02/07/2023]
|
19
|
Stirm M, Fonteyne LM, Shashikadze B, Stöckl JB, Kurome M, Keßler B, Zakhartchenko V, Kemter E, Blum H, Arnold GJ, Matiasek K, Wanke R, Wurst W, Nagashima H, Knieling F, Walter MC, Kupatt C, Fröhlich T, Klymiuk N, Blutke A, Wolf E. Pig models for Duchenne muscular dystrophy – from disease mechanisms to validation of new diagnostic and therapeutic concepts. Neuromuscul Disord 2022; 32:543-556. [DOI: 10.1016/j.nmd.2022.04.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/22/2022] [Accepted: 04/22/2022] [Indexed: 12/13/2022]
|
20
|
Podkalicka P, Mucha O, Kaziród K, Szade K, Stępniewski J, Ivanishchuk L, Hirao H, Pośpiech E, Józkowicz A, Kupiec-Weglinski JW, Dulak J, Łoboda A. miR-378 affects metabolic disturbances in the mdx model of Duchenne muscular dystrophy. Sci Rep 2022; 12:3945. [PMID: 35273230 PMCID: PMC8913680 DOI: 10.1038/s41598-022-07868-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 02/22/2022] [Indexed: 02/08/2023] Open
Abstract
Although Duchenne muscular dystrophy (DMD) primarily affects muscle tissues, the alterations to systemic metabolism manifested in DMD patients contribute to the severe phenotype of this fatal disorder. We propose that microRNA-378a (miR-378) alters carbohydrate and lipid metabolism in dystrophic mdx mice. In our study, we utilized double knockout animals which lacked both dystrophin and miR-378 (mdx/miR-378-/-). RNA sequencing of the liver identified 561 and 194 differentially expressed genes that distinguished mdx versus wild-type (WT) and mdx/miR-378-/- versus mdx counterparts, respectively. Bioinformatics analysis predicted, among others, carbohydrate metabolism disorder in dystrophic mice, as functionally proven by impaired glucose tolerance and insulin sensitivity. The lack of miR-378 in mdx animals mitigated those effects with a faster glucose clearance in a glucose tolerance test (GTT) and normalization of liver glycogen levels. The absence of miR-378 also restored the expression of genes regulating lipid homeostasis, such as Acly, Fasn, Gpam, Pnpla3, and Scd1. In conclusion, we report for the first time that miR-378 loss results in increased systemic metabolism of mdx mice. Together with our previous finding, demonstrating alleviation of the muscle-related symptoms of DMD, we propose that the inhibition of miR-378 may represent a new strategy to attenuate the multifaceted symptoms of DMD.
Collapse
Affiliation(s)
- Paulina Podkalicka
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, 30-387, Kraków, Poland
| | - Olga Mucha
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, 30-387, Kraków, Poland
| | - Katarzyna Kaziród
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, 30-387, Kraków, Poland
| | - Krzysztof Szade
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, 30-387, Kraków, Poland
| | - Jacek Stępniewski
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, 30-387, Kraków, Poland
| | - Liudmyla Ivanishchuk
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, 30-387, Kraków, Poland
| | - Hirofumi Hirao
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Ewelina Pośpiech
- Malopolska Centre of Biotechnology, Jagiellonian University in Krakow, 30-387, Kraków, Poland
| | - Alicja Józkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, 30-387, Kraków, Poland
| | - Jerzy W Kupiec-Weglinski
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, 30-387, Kraków, Poland
| | - Agnieszka Łoboda
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, 30-387, Kraków, Poland.
| |
Collapse
|
21
|
Beck EA, Healey HM, Small CM, Currey MC, Desvignes T, Cresko WA, Postlethwait JH. Advancing human disease research with fish evolutionary mutant models. Trends Genet 2022; 38:22-44. [PMID: 34334238 PMCID: PMC8678158 DOI: 10.1016/j.tig.2021.07.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 01/03/2023]
Abstract
Model organism research is essential to understand disease mechanisms. However, laboratory-induced genetic models can lack genetic variation and often fail to mimic the spectrum of disease severity. Evolutionary mutant models (EMMs) are species with evolved phenotypes that mimic human disease. EMMs complement traditional laboratory models by providing unique avenues to study gene-by-environment interactions, modular mutations in noncoding regions, and their evolved compensations. EMMs have improved our understanding of complex diseases, including cancer, diabetes, and aging, and illuminated mechanisms in many organs. Rapid advancements of sequencing and genome-editing technologies have catapulted the utility of EMMs, particularly in fish. Fish are the most diverse group of vertebrates, exhibiting a kaleidoscope of specialized phenotypes, many that would be pathogenic in humans but are adaptive in the species' specialized habitat. Importantly, evolved compensations can suggest avenues for novel disease therapies. This review summarizes current research using fish EMMs to advance our understanding of human disease.
Collapse
Affiliation(s)
- Emily A Beck
- Data Science, University of Oregon, Eugene, OR 97403, USA; Institute of Ecology and Evolution, University of Oregon, Eugene, OR 97403, USA.
| | - Hope M Healey
- Institute of Ecology and Evolution, University of Oregon, Eugene, OR 97403, USA
| | - Clayton M Small
- Data Science, University of Oregon, Eugene, OR 97403, USA; Institute of Ecology and Evolution, University of Oregon, Eugene, OR 97403, USA
| | - Mark C Currey
- Institute of Ecology and Evolution, University of Oregon, Eugene, OR 97403, USA
| | - Thomas Desvignes
- Institute of Neuroscience, University of Oregon, Eugene, OR 97403, USA
| | - William A Cresko
- Data Science, University of Oregon, Eugene, OR 97403, USA; Institute of Ecology and Evolution, University of Oregon, Eugene, OR 97403, USA
| | | |
Collapse
|
22
|
Ismail MK, Shrestha S. Gastrointestinal Complications of Neuromuscular Disorders. Neuromuscul Disord 2022. [DOI: 10.1016/b978-0-323-71317-7.00004-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
23
|
Stirm M, Fonteyne LM, Shashikadze B, Lindner M, Chirivi M, Lange A, Kaufhold C, Mayer C, Medugorac I, Kessler B, Kurome M, Zakhartchenko V, Hinrichs A, Kemter E, Krause S, Wanke R, Arnold GJ, Wess G, Nagashima H, de Angelis MH, Flenkenthaler F, Kobelke LA, Bearzi C, Rizzi R, Bähr A, Reese S, Matiasek K, Walter MC, Kupatt C, Ziegler S, Bartenstein P, Fröhlich T, Klymiuk N, Blutke A, Wolf E. A scalable, clinically severe pig model for Duchenne muscular dystrophy. Dis Model Mech 2021; 14:273744. [PMID: 34796900 PMCID: PMC8688409 DOI: 10.1242/dmm.049285] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 11/11/2021] [Indexed: 11/20/2022] Open
Abstract
Large animal models for Duchenne muscular dystrophy (DMD) are crucial for evaluation of diagnostic procedures and treatment strategies. Pigs cloned from male cells lacking DMD exon 52 (DMDΔ52) resemble molecular, clinical and pathological hallmarks of DMD, but die before sexual maturity and cannot be propagated by breeding. Therefore, we generated female DMD+/- carriers. A single founder animal had 11 litters with 29 DMDY/-, 34 DMD+/- as well as 36 male and 29 female wild-type offspring. Breeding with F1 and F2 DMD+/- carriers resulted in additional 114 DMDY/- piglets. With intensive neonatal management, the majority survived for 3-4 months, providing statistically relevant cohorts for experimental studies. Pathological investigations and proteome studies of skeletal muscles and myocardium confirmed the resemblance of human disease mechanisms. Importantly, DMDY/- pigs reveal progressive myocardial fibrosis and increased expression of connexin-43, associated with significantly reduced left ventricular ejection fraction already at age 3 months. Furthermore, behavioral tests provided evidence for impaired cognitive ability. Our breeding cohort of DMDΔ52 pigs and standardized tissue repositories provide important resources for studying DMD disease mechanisms and for testing novel treatment strategies.
Collapse
Affiliation(s)
- Michael Stirm
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models (CiMM), LMU Munich, Munich, Germany
| | - Lina Marie Fonteyne
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models (CiMM), LMU Munich, Munich, Germany
| | - Bachuki Shashikadze
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich, Germany
| | - Magdalena Lindner
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Maila Chirivi
- Fondazione Istituto Nazionale di Genetica Molecolare, Milan, Italy.,Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Andreas Lange
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models (CiMM), LMU Munich, Munich, Germany
| | - Clara Kaufhold
- Institute of Veterinary Pathology, Center for Clinical Veterinary Medicine, LMU Munich, Munich, Germany
| | - Christian Mayer
- Institute of Veterinary Pathology, Center for Clinical Veterinary Medicine, LMU Munich, Munich, Germany
| | - Ivica Medugorac
- Population Genomics Group, Department of Veterinary Sciences, LMU Munich, Munich, Germany
| | - Barbara Kessler
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models (CiMM), LMU Munich, Munich, Germany
| | - Mayuko Kurome
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models (CiMM), LMU Munich, Munich, Germany
| | - Valeri Zakhartchenko
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models (CiMM), LMU Munich, Munich, Germany
| | - Arne Hinrichs
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models (CiMM), LMU Munich, Munich, Germany
| | - Elisabeth Kemter
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models (CiMM), LMU Munich, Munich, Germany
| | - Sabine Krause
- Friedrich Baur Institute, Department of Neurology, LMU Munich, Munich, Germany
| | - Rüdiger Wanke
- Institute of Veterinary Pathology, Center for Clinical Veterinary Medicine, LMU Munich, Munich, Germany
| | - Georg J Arnold
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich, Germany
| | - Gerhard Wess
- Clinic of Small Animal Medicine, Center for Clinical Veterinary Medicine, LMU Munich, Munich, Germany
| | - Hiroshi Nagashima
- Meiji University International Institute for Bio-Resource Research, Kawasaki, Japan
| | | | - Florian Flenkenthaler
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich, Germany
| | - Levin Arne Kobelke
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich, Germany
| | - Claudia Bearzi
- Fondazione Istituto Nazionale di Genetica Molecolare, Milan, Italy.,Institute of Genetic and Biomedical Research, UOS of Milan, National Research Council (IRGB-CNR), Milan, Italy
| | - Roberto Rizzi
- Fondazione Istituto Nazionale di Genetica Molecolare, Milan, Italy.,Institute for Biomedical Technologies, National Research Council (ITB-CNR), Segrate, Milan, Italy
| | - Andrea Bähr
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich and German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - Sven Reese
- Chair for Anatomy, Histology and Embryology, Department of Veterinary Sciences, LMU Munich, Munich, Germany
| | - Kaspar Matiasek
- Institute of Veterinary Pathology, Center for Clinical Veterinary Medicine, LMU Munich, Munich, Germany
| | - Maggie C Walter
- Friedrich Baur Institute, Department of Neurology, LMU Munich, Munich, Germany
| | - Christian Kupatt
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich and German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - Sibylle Ziegler
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Thomas Fröhlich
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich, Germany
| | - Nikolai Klymiuk
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models (CiMM), LMU Munich, Munich, Germany
| | - Andreas Blutke
- Institute of Experimental Genetics, Helmholtz Zentrum Munich, Neuherberg, Germany
| | - Eckhard Wolf
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models (CiMM), LMU Munich, Munich, Germany.,Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich, Germany
| |
Collapse
|
24
|
Lionarons JM, Hoogland G, Slegers RJ, Steinbusch H, Claessen SMH, Vles JSH. Dystrophin in the Neonatal and Adult Rat Intestine. Life (Basel) 2021; 11:life11111155. [PMID: 34833031 PMCID: PMC8622973 DOI: 10.3390/life11111155] [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: 08/01/2021] [Revised: 10/25/2021] [Accepted: 10/27/2021] [Indexed: 11/17/2022] Open
Abstract
Background: Gastrointestinal (GI) complaints are frequently noted in aging dystrophinopathy patients, yet their underlying molecular mechanisms are largely unknown. As dystrophin protein isoform 71 (Dp71) is particularly implicated in the development of smooth muscle cells, we evaluated its distribution in the neonatal and adult rat intestine in this study. Methods: Dp71 expression levels were assessed in the proximal (duodenum, jejunum and ileum) and distal (caecum, colon and rectum) intestine by Western blotting and qPCR. In addition, the cellular distribution of total Dp was evaluated in the duodenum and colon by immunohistochemical colocalization studies with alpha-smooth muscle actin (aSMA), Hu RNA binding proteins C and D (HuC/HuD) for neurons and vimentin (VIM) for interstitial cells. Results: In neonatal and adult rats, the distal intestine expressed 2.5 times more Dp71 protein than the proximal part (p < 0.01). This regional difference was not observed in Dp71 mRNA. During both stages, Dp-immunoreactivity was predominant in the muscularis propria, where it co-localized with aSMA and HuC/HuD. Conclusions: In neonatal and adult rats, Dp71 was expressed highest in the distal intestine. Together with the observation that Dp may be expressed by myenteric neurons, this warrants a paradigm shift in the treatment of GI comorbidities.
Collapse
Affiliation(s)
- Judith M. Lionarons
- Department of Neurology, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
- School for Mental Health and Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (R.J.S.); (H.S.); (S.M.H.C.); (J.S.H.V.)
- Correspondence: (J.M.L.); (G.H.); Tel.: +31-(0)43-3875058 (J.M.L.); +31-(0)43-3881024 (G.H.)
| | - Govert Hoogland
- School for Mental Health and Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (R.J.S.); (H.S.); (S.M.H.C.); (J.S.H.V.)
- Department of Neurosurgery, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
- Correspondence: (J.M.L.); (G.H.); Tel.: +31-(0)43-3875058 (J.M.L.); +31-(0)43-3881024 (G.H.)
| | - Rutger J. Slegers
- School for Mental Health and Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (R.J.S.); (H.S.); (S.M.H.C.); (J.S.H.V.)
| | - Hellen Steinbusch
- School for Mental Health and Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (R.J.S.); (H.S.); (S.M.H.C.); (J.S.H.V.)
| | - Sandra M. H. Claessen
- School for Mental Health and Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (R.J.S.); (H.S.); (S.M.H.C.); (J.S.H.V.)
| | - Johan S. H. Vles
- School for Mental Health and Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (R.J.S.); (H.S.); (S.M.H.C.); (J.S.H.V.)
| |
Collapse
|
25
|
Ohlendieck K, Swandulla D. Complexity of skeletal muscle degeneration: multi-systems pathophysiology and organ crosstalk in dystrophinopathy. Pflugers Arch 2021; 473:1813-1839. [PMID: 34553265 PMCID: PMC8599371 DOI: 10.1007/s00424-021-02623-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 02/07/2023]
Abstract
Duchenne muscular dystrophy is a highly progressive muscle wasting disorder due to primary abnormalities in one of the largest genes in the human genome, the DMD gene, which encodes various tissue-specific isoforms of the protein dystrophin. Although dystrophinopathies are classified as primary neuromuscular disorders, the body-wide abnormalities that are associated with this disorder and the occurrence of organ crosstalk suggest that a multi-systems pathophysiological view should be taken for a better overall understanding of the complex aetiology of X-linked muscular dystrophy. This article reviews the molecular and cellular effects of deficiency in dystrophin isoforms in relation to voluntary striated muscles, the cardio-respiratory system, the kidney, the liver, the gastrointestinal tract, the nervous system and the immune system. Based on the establishment of comprehensive biomarker signatures of X-linked muscular dystrophy using large-scale screening of both patient specimens and genetic animal models, this article also discusses the potential usefulness of novel disease markers for more inclusive approaches to differential diagnosis, prognosis and therapy monitoring that also take into account multi-systems aspects of dystrophinopathy. Current therapeutic approaches to combat muscular dystrophy are summarised.
Collapse
Affiliation(s)
- Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, Co. Kildare, Maynooth, W23F2H6, Ireland.
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Co. Kildare, Maynooth, W23F2H6, Ireland.
| | - Dieter Swandulla
- Institute of Physiology, University of Bonn, 53115, Bonn, Germany.
| |
Collapse
|
26
|
Nitric Oxide: From Gastric Motility to Gastric Dysmotility. Int J Mol Sci 2021; 22:ijms22189990. [PMID: 34576155 PMCID: PMC8470306 DOI: 10.3390/ijms22189990] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/11/2021] [Accepted: 09/13/2021] [Indexed: 12/27/2022] Open
Abstract
It is known that nitric oxide (NO) plays a key physiological role in the control of gastrointestinal (GI) motor phenomena. In this respect, NO is considered as the main non-adrenergic, non-cholinergic (NANC) inhibitory neurotransmitter responsible for smooth muscle relaxation. Moreover, many substances (including hormones) have been reported to modulate NO production leading to changes in motor responses, further underlying the importance of this molecule in the control of GI motility. An impaired NO production/release has indeed been reported to be implicated in some GI dysmotility. In this article we wanted to focus on the influence of NO on gastric motility by summarizing knowledge regarding its role in both physiological and pathological conditions. The main role of NO on regulating gastric smooth muscle motor responses, with particular reference to NO synthases expression and signaling pathways, is discussed. A deeper knowledge of nitrergic mechanisms is important for a better understanding of their involvement in gastric pathophysiological conditions of hypo- or hyper-motility states and for future therapeutic approaches. A possible role of substances which, by interfering with NO production, could prove useful in managing such motor disorders has been advanced.
Collapse
|
27
|
Lionarons JM, de Groot IJM, Fock JM, Klinkenberg S, Vrijens DMJ, Vreugdenhil ACE, Medici-van den Herik EG, Cuppen I, Jaeger B, Niks EH, Hoogerhuis R, Platte-van Attekum N, Feron FJM, Faber CG, Hendriksen JGM, Vles JSH. Prevalence of Bladder and Bowel Dysfunction in Duchenne Muscular Dystrophy Using the Childhood Bladder and Bowel Dysfunction Questionnaire. Life (Basel) 2021; 11:life11080772. [PMID: 34440515 PMCID: PMC8399211 DOI: 10.3390/life11080772] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/29/2021] [Accepted: 07/27/2021] [Indexed: 12/27/2022] Open
Abstract
Introduction: Lower urinary tract symptoms (LUTS) and gastrointestinal (GI) problems are common in Duchenne muscular dystrophy (DMD), but not systematically assessed in regular care. We aimed to determine the prevalence of bladder and bowel dysfunction (BBD) in DMD patients compared with healthy controls (HC). Methods: The Childhood Bladder and Bowel Dysfunction Questionnaire (CBBDQ) based on the International Rome III criteria and the International Children’s Continence Society was filled out by 57 DMD patients and 56 HC. Additionally, possible associations of BBD with, for example, medication use or quality of life were evaluated in an additional questionnaire developed by experts. Results: In 74% of patients versus 56% of HC ≥ 1 LUTS (n.s.) were reported, 68% of patients versus 39% of HC reported ≥1 bowel symptom (p = 0.002) and 53% of patients versus 30% of HC reported combined LUTS and bowel symptoms (p = 0.019). A negative impact of BBD on daily life functioning was reported by 42% of patients. Conclusions: These data underscore that standard screening for BBD is needed and that the CBBDQ could be of added value to optimize DMD care.
Collapse
Affiliation(s)
- Judith M. Lionarons
- Department of Neurology, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands; (S.K.); (C.G.F.)
- School for Mental Health and Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands;
- Correspondence: ; Tel.: +31-(0)43-3875058
| | - Imelda J. M. de Groot
- Department of Rehabilitation Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
- Duchenne Center Netherlands, 2333 ZA Leiden, The Netherlands; (E.H.N.); (J.G.M.H.)
| | - Johanna M. Fock
- Department of Neurology, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands;
| | - Sylvia Klinkenberg
- Department of Neurology, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands; (S.K.); (C.G.F.)
| | - Desiree M. J. Vrijens
- Department of Urology, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands;
| | - Anita C. E. Vreugdenhil
- Department of Pediatrics, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands;
- School of Nutrition and Translational Research in Metabolism, Maastricht University, 6229 ER Maastricht, The Netherlands
| | | | - Inge Cuppen
- Department of Neurology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands;
| | - Bregje Jaeger
- Department of Neurology, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands;
| | - Erik H. Niks
- Duchenne Center Netherlands, 2333 ZA Leiden, The Netherlands; (E.H.N.); (J.G.M.H.)
- Department of Neurology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Rinske Hoogerhuis
- Youth Healthcare Center South Limburg, 6411 TE Heerlen, The Netherlands; (R.H.); (N.P.-v.A.)
| | | | - Frans J. M. Feron
- Department of Social Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands;
| | - Catharina G. Faber
- Department of Neurology, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands; (S.K.); (C.G.F.)
- School for Mental Health and Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands;
| | - Jos G. M. Hendriksen
- Duchenne Center Netherlands, 2333 ZA Leiden, The Netherlands; (E.H.N.); (J.G.M.H.)
- Center for Neurological Learning Disabilities, Kempenhaeghe, 5591 VE Heeze, The Netherlands
| | - Johan S. H. Vles
- School for Mental Health and Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands;
| |
Collapse
|
28
|
Mosiello G, Safder S, Marshall D, Rolle U, Benninga MA. Neurogenic Bowel Dysfunction in Children and Adolescents. J Clin Med 2021; 10:1669. [PMID: 33924675 PMCID: PMC8069792 DOI: 10.3390/jcm10081669] [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: 02/21/2021] [Revised: 04/06/2021] [Accepted: 04/08/2021] [Indexed: 01/07/2023] Open
Abstract
Neurogenic/neuropathic bowel dysfunction (NBD) is common in children who are affected by congenital and acquired neurological disease, and negatively impacts quality of life. In the past, NBD received less attention than neurogenic bladder, generally being considered only in spina bifida (the most common cause of pediatric NBD). Many methods of conservative and medical management of NBD are reported, including relatively recently Transanal Irrigation (TAI). Based on the literature and personal experience, an expert group (pediatric urologists/surgeons/gastroenterologists with specific experience in NBD) focused on NBD in children and adolescents. A statement document was created using a modified Delphi method. The range of causes of pediatric NBD are discussed in this paper. The various therapeutic approaches are presented to improve clinical management. The population of children and adolescents with NBD is increasing, due both to the higher survival rate and better diagnosis. While NBD is relatively predictable in producing either constipation or fecal incontinence, or both, its various effects on each patient will depend on a wide range of underlying causes and accompanying comorbidities. For this reason, management of NBD should be tailored individually with a combined multidisciplinary therapy appropriate for the status of the affected child and caregivers.
Collapse
Affiliation(s)
- Giovanni Mosiello
- Department of Surgery, Division of Urology, Bambino Gesù Pediatric and Research Hospital, 00165 Rome, Italy
| | - Shaista Safder
- College of Medicine, Center for Digestive, Health and Nutrition, Arnold Palmer Hospital for Children, Orlando, FL 32806, USA;
| | - David Marshall
- Department of Pediatric Surgery and Pediatric Urology, Royal Belfast Hospital for Sick Children, Belfast BT97AB, UK;
| | - Udo Rolle
- Department of Pediatric Surgery and Pediatric Urology, Goethe-University Frankfurt, 60596 Frankfurt, Germany;
| | - Marc A. Benninga
- Department of Pediatric Gastroenterology, Hepatology and Nutrition, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| |
Collapse
|
29
|
Dowling P, Gargan S, Zweyer M, Sabir H, Henry M, Meleady P, Swandulla D, Ohlendieck K. Proteomic profiling of the interface between the stomach wall and the pancreas in dystrophinopathy. Eur J Transl Myol 2021; 31. [PMID: 33709651 PMCID: PMC8056161 DOI: 10.4081/ejtm.2021.9627] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 02/03/2021] [Indexed: 02/08/2023] Open
Abstract
The neuromuscular disorder Duchenne muscular dystrophy is a multi-systemic disease that is caused by a primary abnormality in the X-chromosomal Dmd gene. Although progressive skeletal muscle wasting and cardio-respiratory complications are the most serious symptoms that are directly linked to the almost complete loss of the membrane cytoskeletal protein dystrophin, dystrophic patients also suffer from gastrointestinal dysfunction. In order to determine whether proteome-wide changes potentially occur in the gastrointestinal system due to dystrophin deficiency, total tissue extracts from the interface between the stomach wall and the pancreas of the mdx-4cv model of dystrophinopathy were analysed by mass spectrometry. Following the proteomic establishment of both smooth muscle markers of the gastrointestinal system and key enzymes of the pancreas, core members of the dystrophin-glycoprotein complex, including dystrophin, dystroglycans, sarcoglycans, dystrobrevins and syntrophins were identified in this tissue preparation. Comparative proteomics revealed a drastic reduction in dystrophin, sarcoglycan, dystroglycan, laminin, titin and filamin suggesting loss of cytoskeletal integrity in mdx-4cv smooth muscles. A concomitant increase in various mitochondrial enzymes is indicative of metabolic disturbances. These findings agree with abnormal gastrointestinal function in dystrophinopathy.
Collapse
Affiliation(s)
- Paul Dowling
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland; Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare.
| | - Stephen Gargan
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland; Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare.
| | - Margit Zweyer
- Department of Neonatology and Paediatric Intensive Care, Children's Hospital, University of Bonn, Bonn.
| | - Hemmen Sabir
- Department of Neonatology and Paediatric Intensive Care, Children's Hospital, University of Bonn, Bonn.
| | - Michael Henry
- National Institute for Cellular Biotechnology, Dublin City University, Dublin 9.
| | - Paula Meleady
- National Institute for Cellular Biotechnology, Dublin City University, Dublin 9.
| | | | - Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland; Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare.
| |
Collapse
|
30
|
Hossain SF, Huang M, Ono N, Morita A, Kanaya S, Altaf-Ul-Amin M. Development of a biomarker database toward performing disease classification and finding disease interrelations. Database (Oxford) 2021; 2021:baab011. [PMID: 33705530 PMCID: PMC7951048 DOI: 10.1093/database/baab011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 02/19/2021] [Accepted: 02/25/2021] [Indexed: 12/11/2022]
Abstract
A biomarker is a measurable indicator of a disease or abnormal state of a body that plays an important role in disease diagnosis, prognosis and treatment. The biomarker has become a significant topic due to its versatile usage in the medical field and in rapid detection of the presence or severity of some diseases. The volume of biomarker data is rapidly increasing and the identified data are scattered. To provide comprehensive information, the explosively growing data need to be recorded in a single platform. There is no open-source freely available comprehensive online biomarker database. To fulfill this purpose, we have developed a human biomarker database as part of the KNApSAcK family databases which contain a vast quantity of information on the relationships between biomarkers and diseases. We have classified the diseases into 18 disease classes, mostly according to the National Center for Biotechnology Information definitions. Apart from this database development, we also have performed disease classification by separately using protein and metabolite biomarkers based on the network clustering algorithm DPClusO and hierarchical clustering. Finally, we reached a conclusion about the relationships among the disease classes. The human biomarker database can be accessed online and the inter-disease relationships may be helpful in understanding the molecular mechanisms of diseases. To our knowledge, this is one of the first approaches to classify diseases based on biomarkers. Database URL: http://www.knapsackfamily.com/Biomarker/top.php.
Collapse
Affiliation(s)
- Shaikh Farhad Hossain
- Computational Systems Biology Lab, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), 8916-5, Takayama, Ikoma, Nara 630-0192, Japan
| | - Ming Huang
- Computational Systems Biology Lab, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), 8916-5, Takayama, Ikoma, Nara 630-0192, Japan
| | - Naoaki Ono
- Computational Systems Biology Lab, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), 8916-5, Takayama, Ikoma, Nara 630-0192, Japan
| | - Aki Morita
- Computational Systems Biology Lab, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), 8916-5, Takayama, Ikoma, Nara 630-0192, Japan
| | - Shigehiko Kanaya
- Computational Systems Biology Lab, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), 8916-5, Takayama, Ikoma, Nara 630-0192, Japan
| | - Md Altaf-Ul-Amin
- Computational Systems Biology Lab, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), 8916-5, Takayama, Ikoma, Nara 630-0192, Japan
| |
Collapse
|
31
|
Dowling P, Gargan S, Zweyer M, Sabir H, Henry M, Meleady P, Swandulla D, Ohlendieck K. Proteomic profiling of the interface between the stomach wall and the pancreas in dystrophinopathy. Eur J Transl Myol 2021. [DOI: 10.4081/ejtm.2020.9627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The neuromuscular disorder Duchenne muscular dystrophy is a multi-systemic disease that is caused by a primary abnormality in the X-chromosomal Dmd gene. Although progressive skeletal muscle wasting and cardio-respiratory complications are the most serious symptoms that are directly linked to the almost complete loss of the membrane cytoskeletal protein dystrophin, dystrophic patients also suffer from gastrointestinal dysfunction. In order to determine whether proteome-wide changes potentially occur in the gastrointestinal system due to dystrophin deficiency, total tissue extracts from the interface between the stomach wall and the pancreas of the mdx-4cv model of dystrophinopathy were analysed by mass spectrometry. Following the proteomic establishment of both smooth muscle markers of the gastrointestinal system and key enzymes of the pancreas, core members of the dystrophin-glycoprotein complex, including dystrophin, dystroglycans, sarcoglycans, dystrobrevins and syntrophins were identified in this tissue preparation. Comparative proteomics revealed a drastic reduction in dystrophin, sarcoglycan, dystroglycan, laminin, titin and filamin suggesting loss of cytoskeletal integrity in mdx-4cv smooth muscles. A concomitant increase in various mitochondrial enzymes is indicative of metabolic disturbances. These findings agree with abnormal gastrointestinal function in dystrophinopathy.
Collapse
|
32
|
Singh K, Randhwa G, Salloum FN, Grider JR, Murthy KS. Decreased smooth muscle function, peristaltic activity, and gastrointestinal transit in dystrophic (mdx) mice. Neurogastroenterol Motil 2021; 33:e13968. [PMID: 32789934 DOI: 10.1111/nmo.13968] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 07/21/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is characterized by the lack of dystrophin in skeletal, cardiac, and smooth muscle. Slow colonic transit and constipation are common in DMD patients and animal models of DMD. However, the cause of this hypocontractility and the expression of contractile proteins in smooth muscle are unknown. The aim of the study was to investigate the expression of contractile proteins in the colonic smooth muscle and the function of the colon in control and mdx mice. METHODS Muscle contraction was measured in muscle strips and isolated muscle cells. Peristaltic activity was measured in ex vivo preparations by spatiotemporal mapping, and gastrointestinal (GI) transit in vivo was measured by the distribution of fluorescent marker along the intestine and colon. mRNA expression of contractile proteins smoothelin, caldesmon, calponin, and tropomyosin was measured by qRT-PCR. RESULTS Expression of mRNA for contractile proteins was decreased in colonic smooth muscle of mdx mice compared with control. Contraction in response to acetylcholine and KCl was decreased in colonic muscle strips and in isolated muscle cells of mdx mice. Distension of ex vivo colons with Krebs buffer induced peristalsis in both control and mdx mice; however, significantly fewer full peristaltic waves were recorded in the colons of mdx mice. GI transit was also inhibited in mdx mice. CONCLUSION AND INFERENCES The data indicate that the lack of dystrophin causes decrease in colonic smooth muscle contractility, peristalsis, and GI transit and provides the basis for analysis of mechanisms involved in smooth muscle dysfunction in DMD.
Collapse
Affiliation(s)
- Kulpreet Singh
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, USA
| | - Gurpreet Randhwa
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, USA
| | - Fadi N Salloum
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, USA
| | - John R Grider
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, USA
| | - Karnam S Murthy
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
33
|
Quinlivan R, Messer B, Murphy P, Astin R, Mukherjee R, Khan J, Emmanuel A, Wong S, Kulshresha R, Willis T, Pattni J, Willis D, Morgan A, Savvatis K, Keen R, Bourke J, Marini Bettolo C, Hewamadduma C. Adult North Star Network (ANSN): Consensus Guideline For The Standard Of Care Of Adults With Duchenne Muscular Dystrophy. J Neuromuscul Dis 2021; 8:899-926. [PMID: 34511509 PMCID: PMC8673515 DOI: 10.3233/jnd-200609] [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] [Indexed: 12/28/2022]
Abstract
There are growing numbers of adults with Duchenne Muscular Dystrophy living well into their fourth decade. These patients have complex medical needs that to date have not been addressed in the International standards of care. We sought to create a consensus based standard of care through a series of multi-disciplinary workshops with specialists from a wide range of clinical areas: Neurology, Cardiology, Respiratory Medicine, Gastroenterology, Endocrinology, Palliative Care Medicine, Rehabilitation, Renal, Anaesthetics and Clinical Psychology. Detailed reports of evidence reviewed and the consensus building process were produced following each workshop and condensed into this final document which was approved by all members of the Adult North Star Network including service users. The aim of this document is to provide a framework to improve clinical services and multi-disciplinary care for adults living with Duchenne Muscular Dystrophy.
Collapse
Affiliation(s)
- R. Quinlivan
- MRC Centre for Neuromuscular Disease, UCL Institute of Neurology, National Hospital for Neurology and Neurosurgery, London, UK
| | - B. Messer
- Newcastle-upon-Tyne Hospitals NHS Foundation Trust, Newcastle, UK
| | - P. Murphy
- Lane Fox Unit, Guy’s and St Thomas’ Foundation Trust, London, UK
| | - R. Astin
- MRC Centre for Neuromuscular Disease, UCL Institute of Neurology, National Hospital for Neurology and Neurosurgery, London, UK
| | - R. Mukherjee
- Heart of England NHS Foundation Trust, Birmingham, UK
| | - J. Khan
- MRC Centre for Neuromuscular Disease, UCL Institute of Neurology, National Hospital for Neurology and Neurosurgery, London, UK
| | - A. Emmanuel
- MRC Centre for Neuromuscular Disease, UCL Institute of Neurology, National Hospital for Neurology and Neurosurgery, London, UK
| | - S.C. Wong
- University of Glasgow, Royal Hospital for Children, Glasgow, UK
| | - R. Kulshresha
- Robert Jones and Agnes Hunt Foundation NHS Trust, Oswestry, UK
| | - T. Willis
- Robert Jones and Agnes Hunt Foundation NHS Trust, Oswestry, UK
| | - J. Pattni
- MRC Centre for Neuromuscular Disease, UCL Institute of Neurology, National Hospital for Neurology and Neurosurgery, London, UK
| | - D. Willis
- Shrewsbury and Telford NHS Trust, Shropshire, UK
| | - A. Morgan
- South West Neuromuscular Operational Delivery Network, Bristol, UK
| | - K. Savvatis
- MRC Centre for Neuromuscular Disease, UCL Institute of Neurology, National Hospital for Neurology and Neurosurgery, London, UK
- St Bartholomew’s Hospital and Royal London NHS Trust, London UK
| | - R. Keen
- Royal National Orthopaedic Hospital, Stanmore, UK
| | - J. Bourke
- Newcastle-upon-Tyne Hospitals NHS Foundation Trust, Newcastle, UK
| | | | - C. Hewamadduma
- Academic Neurology Department, Sheffield Teaching Hospitals Foundation Trust and Sheffield Institute for Translational Neurosciences (SITRAN), University of Sheffield, Sheffield, UK
| | - on behalf of the ANSN
- MRC Centre for Neuromuscular Disease, UCL Institute of Neurology, National Hospital for Neurology and Neurosurgery, London, UK
- Newcastle-upon-Tyne Hospitals NHS Foundation Trust, Newcastle, UK
- Lane Fox Unit, Guy’s and St Thomas’ Foundation Trust, London, UK
- Heart of England NHS Foundation Trust, Birmingham, UK
- University of Glasgow, Royal Hospital for Children, Glasgow, UK
- Robert Jones and Agnes Hunt Foundation NHS Trust, Oswestry, UK
- Shrewsbury and Telford NHS Trust, Shropshire, UK
- South West Neuromuscular Operational Delivery Network, Bristol, UK
- St Bartholomew’s Hospital and Royal London NHS Trust, London UK
- Royal National Orthopaedic Hospital, Stanmore, UK
- Academic Neurology Department, Sheffield Teaching Hospitals Foundation Trust and Sheffield Institute for Translational Neurosciences (SITRAN), University of Sheffield, Sheffield, UK
| |
Collapse
|
34
|
Lee JW, Oh HJ, Choi WA, Kim DJ, Kang SW. Relationship between Eating and Digestive Symptoms and Respiratory Function in Advanced Duchenne Muscular Dystrophy Patients. J Neuromuscul Dis 2020; 7:101-107. [PMID: 31903995 PMCID: PMC7175944 DOI: 10.3233/jnd-190435] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background: Duchenne muscular dystrophy (DMD) patients can have various issues that affect their quality of life, including eating and digestive conditions. Objective: We sought to identify the relationship between respiratory function and various eating and digestion related symptoms in patients with advanced Duchenne muscular dystrophy (DMD). Methods: Eating and digestive symptoms, including loss of appetite, nausea, vomiting, diarrhea, constipation, swallowing difficulty, mastication difficulty, early satiety, and aspiration, were evaluated among patients with advanced DMD who were nonambulatory and required noninvasive mechanical ventilatory support. In addition, various respiratory function parameters were measured, including forced vital capacity (FVC), maximal insufflation capacity (MIC), peak cough flow (PCF), assisted PCF (APCF), maximal inspiratory pressure (MIP), and maximal expiratory pressure (MEP). We then analyzed the relationship between gastrointestinal symptoms and respiratory function parameters. Results: A total of 180 patients (age, 22.3±5.0 years) were included in the analysis. Loss of appetite and early satiety showed no correlation with any of the respiratory function parameters. Constipation was correlated with MEP; swallowing difficulty was correlated with MIC, APCF, MIP and MEP; and mastication difficulty was correlated with FVC, PCF, APCF, MIP, and MEP. Notably, age did not correlate with any gastrointestinal symptoms. Conclusions: Eating and digestive symptoms are more closely correlated with respiratory function than with age in patients with DMD. We think this correlation is mainly caused by the skeletal muscle strength, which is major determinant of both digestive and respiratory function.
Collapse
Affiliation(s)
- Jang Woo Lee
- Department of Physical Medicine and Rehabilitation, National Health Insurance Service Ilsan Hospital, Goyang-si, Gyeonggi-do, Korea.,Yonsei University Graduate School of Medicine, Seoul, Korea
| | - Hyun Jun Oh
- Department of Rehabilitation Medicine and Rehabilitation Institute of Neuromuscular Disease, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Won Ah Choi
- Department of Rehabilitation Medicine and Rehabilitation Institute of Neuromuscular Disease, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.,Pulmonary Rehabilitation Center, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Dong Jin Kim
- Department of Rehabilitation Medicine, SRC Rehabilitation Hospital, Gwangju-si, Gyeonggi-do, Korea
| | - Seong-Woong Kang
- Department of Rehabilitation Medicine and Rehabilitation Institute of Neuromuscular Disease, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.,Pulmonary Rehabilitation Center, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.,Yonsei University Graduate School of Medicine, Seoul, Korea
| |
Collapse
|
35
|
Kovacic K, Elfar W, Rosen JM, Yacob D, Raynor J, Mostamand S, Punati J, Fortunato JE, Saps M. Update on pediatric gastroparesis: A review of the published literature and recommendations for future research. Neurogastroenterol Motil 2020; 32:e13780. [PMID: 31854057 DOI: 10.1111/nmo.13780] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 11/11/2019] [Accepted: 11/29/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Due to scarcity of scientific literature on pediatric gastroparesis, there is a need to summarize current evidence and identify areas requiring further research. The aim of this study was to provide an evidence-based review of the available literature on the prevalence, pathogenesis, clinical presentation, diagnosis, treatment, and outcomes of pediatric gastroparesis. METHODS A search of the literature was performed using the Preferred Reporting Items for Systematic Reviews and Meta-analyses guidelines with the following databases: PubMed, EMBASE, Cochrane Database of Systematic Reviews, Database of Abstracts of Reviews of Effects, Cochrane Central Register of Controlled Trials, and Web of Science. Two independent reviewers screened abstracts for eligibility. KEY RESULTS Our search yielded 1085 original publications, 135 of which met inclusion criteria. Most articles were of retrospective study design. Only 12 randomized controlled trials were identified, all of which were in infants. The prevalence of pediatric gastroparesis is unknown. Gastroparesis may be suspected based on clinical symptoms although these are often non-specific. The 4-hour nuclear scintigraphy scan remains gold standard for diagnosis despite lack of pediatric normative comparison data. Therapeutic approaches include dietary modifications, prokinetic drugs, and postpyloric enteral tube feeds. For refractory cases, intrapyloric botulinum toxin and surgical interventions such as gastric electrical stimulation may be warranted. Most interventions still lack rigorous supportive data. CONCLUSIONS Diagnosis and treatment of pediatric gastroparesis are challenging due to paucity of published evidence. Larger and more rigorous clinical trials are necessary to improve outcomes.
Collapse
Affiliation(s)
- Katja Kovacic
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Walaa Elfar
- Division of Gastroenterology and Nutrition, Department of Pediatrics, The Pennsylvania State Melton S. Hershey Medical Center, Hershey, PA, USA
| | - John M Rosen
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, The Children's Mercy Hospital, Kansas City, MO, USA
| | - Desale Yacob
- Division of Gastroenterology, Hepatology, and Nutrition, Nationwide Children's Hospital, Ohio State University, Columbus, OH, USA
| | - Jennifer Raynor
- Edward G. Miner Library, University of Rochester Medical Center, Rochester, NY, USA
| | - Shikib Mostamand
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Jaya Punati
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - John E Fortunato
- Neurointestinal and Motility Program, Section of Pediatric Gastroenterology, Hepatology and Nutrition, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Miguel Saps
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Holtz Children's Hospital, Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
36
|
QTL Mapping of Intestinal Neutrophil Variation in Threespine Stickleback Reveals Possible Gene Targets Connecting Intestinal Inflammation and Systemic Health. G3-GENES GENOMES GENETICS 2020; 10:613-622. [PMID: 31843804 PMCID: PMC7003091 DOI: 10.1534/g3.119.400685] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Selection, via host immunity, is often required to foster beneficial microbial symbionts and suppress deleterious pathogens. In animals, the host immune system is at the center of this relationship. Failed host immune system-microbial interactions can result in a persistent inflammatory response in which the immune system indiscriminately attacks resident microbes, and at times the host cells themselves, leading to diseases such as Ulcerative Colitis, Crohn’s Disease, and Psoriasis. Host genetic variation has been linked to both microbiome diversity and to severity of such inflammatory disease states in humans. However, the microbiome and inflammatory states manifest as quantitative traits, which encompass many genes interacting with one another and the environment. The mechanistic relationships among all of these interacting components are still not clear. Developing natural genetic models of host-microbe interactions is therefore fundamental to understanding the complex genetics of these and other diseases. Threespine stickleback (Gasterosteus aculeatus) fish are a tractable model for attacking this problem because of abundant population-level genetic and phenotypic variation in the gut inflammatory response. Previous work in our laboratory identified genetically divergent stickleback populations exhibiting differences in intestinal neutrophil activity. We took advantage of this diversity to genetically map variation in an emblematic element of gut inflammation - intestinal neutrophil recruitment - using an F2-intercross mapping framework. We identified two regions of the genome associated with increased intestinal inflammation containing several promising candidate genes. Within these regions we found candidates in the Coagulation/Complement System, NFkB and MAPK pathways along with several genes associated with intestinal diseases and neurological diseases commonly accompanying intestinal inflammation as a secondary symptom. These findings highlight the utility of using naturally genetically diverse ‘evolutionary mutant models’ such as threespine stickleback to better understand interactions among host genetic diversity and microbiome variation in health and disease states.
Collapse
|
37
|
Yatsenko AS, Kucherenko MM, Xie Y, Aweida D, Urlaub H, Scheibe RJ, Cohen S, Shcherbata HR. Profiling of the muscle-specific dystroglycan interactome reveals the role of Hippo signaling in muscular dystrophy and age-dependent muscle atrophy. BMC Med 2020; 18:8. [PMID: 31959160 PMCID: PMC6971923 DOI: 10.1186/s12916-019-1478-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 12/05/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Dystroglycanopathies are a group of inherited disorders characterized by vast clinical and genetic heterogeneity and caused by abnormal functioning of the ECM receptor dystroglycan (Dg). Remarkably, among many cases of diagnosed dystroglycanopathies, only a small fraction can be linked directly to mutations in Dg or its regulatory enzymes, implying the involvement of other, not-yet-characterized, Dg-regulating factors. To advance disease diagnostics and develop new treatment strategies, new approaches to find dystroglycanopathy-related factors should be considered. The Dg complex is highly evolutionarily conserved; therefore, model genetic organisms provide excellent systems to address this challenge. In particular, Drosophila is amenable to experiments not feasible in any other system, allowing original insights about the functional interactors of the Dg complex. METHODS To identify new players contributing to dystroglycanopathies, we used Drosophila as a genetic muscular dystrophy model. Using mass spectrometry, we searched for muscle-specific Dg interactors. Next, in silico analyses allowed us to determine their association with diseases and pathological conditions in humans. Using immunohistochemical, biochemical, and genetic interaction approaches followed by the detailed analysis of the muscle tissue architecture, we verified Dg interaction with some of the discovered factors. Analyses of mouse muscles and myocytes were used to test if interactions are conserved in vertebrates. RESULTS The muscle-specific Dg complexome revealed novel components that influence the efficiency of Dg function in the muscles. We identified the closest human homologs for Dg-interacting partners, determined their significant enrichment in disease-associations, and verified some of the newly identified Dg interactions. We found that Dg associates with two components of the mechanosignaling Hippo pathway: the WW domain-containing proteins Kibra and Yorkie. Importantly, this conserved interaction manages adult muscle size and integrity. CONCLUSIONS The results presented in this study provide a new list of muscle-specific Dg interactors, further analysis of which could aid not only in the diagnosis of muscular dystrophies, but also in the development of new therapeutics. To regulate muscle fitness during aging and disease, Dg associates with Kibra and Yorkie and acts as a transmembrane Hippo signaling receptor that transmits extracellular information to intracellular signaling cascades, regulating muscle gene expression.
Collapse
Affiliation(s)
- Andriy S Yatsenko
- Gene Expression and Signaling Group, Institute of Cell Biochemistry, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany
| | - Mariya M Kucherenko
- Max Planck Research Group of Gene Expression and Signaling, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany.,Present Address: Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.,Institute of Physiology, Charité - University Medicine Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Yuanbin Xie
- Max Planck Research Group of Gene Expression and Signaling, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany.,Present Address: University Medical Center, Centre for Anatomy, Institute of Neuroanatomy, Georg-August-University Göttingen, Kreuzbergring 36, 37075, Göttingen, Germany
| | - Dina Aweida
- Faculty of Biology, Technion, 32000, Haifa, Israel
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry Research Group, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany.,Bioanalytics Institute for Clinical Chemistry, University Medical Center Goettingen, Robert Koch Strasse 40, 37075, Göttingen, Germany
| | - Renate J Scheibe
- Gene Expression and Signaling Group, Institute of Cell Biochemistry, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany
| | | | - Halyna R Shcherbata
- Gene Expression and Signaling Group, Institute of Cell Biochemistry, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany. .,Max Planck Research Group of Gene Expression and Signaling, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany.
| |
Collapse
|
38
|
West JD, Galindo CL, Kim K, Shin JJ, Atkinson JB, Macias‐Perez I, Pavliv L, Knollmann BC, Soslow JH, Markham LW, Carrier EJ. Antagonism of the Thromboxane-Prostanoid Receptor as a Potential Therapy for Cardiomyopathy of Muscular Dystrophy. J Am Heart Assoc 2019; 8:e011902. [PMID: 31662020 PMCID: PMC6898850 DOI: 10.1161/jaha.118.011902] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Background Muscular dystrophy (MD) causes a progressive cardiomyopathy characterized by diffuse fibrosis, arrhythmia, heart failure, and early death. Activation of the thromboxane-prostanoid receptor (TPr) increases calcium transients in cardiomyocytes and is proarrhythmic and profibrotic. We hypothesized that TPr activation contributes to the cardiac phenotype of MD, and that TPr antagonism would improve cardiac fibrosis and function in preclinical models of MD. Methods and Results Three different mouse models of MD (mdx/utrn double knockout, second generation mdx/mTR double knockout, and delta-sarcoglycan knockout) were given normal drinking water or water containing 25 mg/kg per day of the TPr antagonist ifetroban, beginning at weaning. After 6 months (10 weeks for mdx/utrn double knockout), mice were evaluated for cardiac and skeletal muscle function before euthanization. There was a 100% survival rate of ifetroban-treated mice to the predetermined end point, compared with 60%, 43%, and 90% for mdx/utrn double knockout, mdx/mTR double knockout, and delta-sarcoglycan knockout mice, respectively. TPr antagonism improved cardiac output in mdx/utrn double knockout and mdx/mTR mice, and normalized fractional shortening, ejection fraction, and other parameters in delta-sarcoglycan knockout mice. Cardiac fibrosis in delta-sarcoglycan knockout was reduced with TPr antagonism, which also normalized cardiac expression of claudin-5 and neuronal nitric oxide synthase proteins and multiple signature genes of Duchenne MD. Conclusions TPr antagonism reduced cardiomyopathy and spontaneous death in mouse models of Duchenne and limb-girdle MD. Based on these studies, ifetroban and other TPr antagonists could be novel therapeutics for treatment of the cardiac phenotype in patients with MD.
Collapse
Affiliation(s)
- James D. West
- Division of Allergy, Pulmonary, and Critical CareVanderbilt University Medical CenterNashvilleTN
| | - Cristi L. Galindo
- Division of CardiologyVanderbilt University Medical CenterNashvilleTN
| | - Kyungsoo Kim
- Division of Clinical PharmacologyVanderbilt University Medical CenterNashvilleTN
| | - John Jonghyun Shin
- Division of Rheumatology and ImmunologyDepartment of MedicineVanderbilt University Medical CenterNashvilleTN
| | - James B. Atkinson
- Department of MedicineDepartment of Pathology, Microbiology, and ImmunologyVanderbilt University Medical CenterNashvilleTN
| | | | - Leo Pavliv
- Cumberland Pharmaceuticals IncNashvilleTN
| | - Bjorn C. Knollmann
- Division of Clinical PharmacologyVanderbilt University Medical CenterNashvilleTN
| | - Jonathan H. Soslow
- Division of Pediatric CardiologyDepartment of PediatricsVanderbilt University Medical CenterNashvilleTN
| | - Larry W. Markham
- Division of CardiologyVanderbilt University Medical CenterNashvilleTN
- Division of Pediatric CardiologyDepartment of PediatricsRiley Hospital for Children and Indiana University School of MedicineIndianapolisIN
| | - Erica J. Carrier
- Division of Allergy, Pulmonary, and Critical CareVanderbilt University Medical CenterNashvilleTN
| |
Collapse
|
39
|
Upper Gastrointestinal Manifestation of Bezoars and the Etiological Factors: A Literature Review. Gastroenterol Res Pract 2019; 2019:5698532. [PMID: 31396274 PMCID: PMC6664490 DOI: 10.1155/2019/5698532] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 04/28/2019] [Accepted: 05/09/2019] [Indexed: 12/19/2022] Open
Abstract
A gastric bezoar is a compact mass of indigestible foreign materials that accumulate and consolidate in the stomach; however, it can be found in other sites of the gastrointestinal tract. The causative manner of this condition is complex and multifactorial. The main purpose of the review was to raise awareness among clinicians, particularly gastroenterologists, that patients with certain risk factors or comorbid conditions are predisposed to gastric bezoar formation. Early diagnosis and prompt intervention are crucial to avoid bezoar-induced complications. Upper gastrointestinal endoscopy is the standard diagnostic and therapeutic method for gastric bezoars. However, for large size bezoars, surgical intervention is needed.
Collapse
|
40
|
Schwarz EI, Bloch KE. Frontiers in Clinical Practice of Long-Term Care of Chronic Ventilatory Failure. Respiration 2019; 98:1-15. [PMID: 31170716 DOI: 10.1159/000499316] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 03/02/2019] [Indexed: 11/19/2022] Open
Abstract
Home mechanical ventilation (HMV) is an effective long-term treatment for chronic hypercapnic respiratory failure. In addition to the established practice of providing HMV for the treatment of chronic ventilatory failure in slowly progressive neuromuscular and chest wall disease, there is accumulating evidence for improvement of quality of life and prolongation of survival by HMV in highly prevalent diseases like chronic obstructive pulmonary disease and ever-increasing obesity hypoventilation syndrome as well as rapidly progressive neuromuscular disease. The key concepts for successful HMV are an experienced team selecting the right patients, timely initiation of adequate ventilation via an appropriate interface, and monitoring effectiveness during regular long-term follow-up. Coaching of patients with chronic respiratory failure on long-term HMV within a dedicated service and collaborations with community services for home care are essential. The current review describes various important practical aspects of HMV that remain frontiers in the implementation of the current knowledge in clinical practice and may help in providing effective HMV to all those in need.
Collapse
Affiliation(s)
- Esther I Schwarz
- Department of Respiratory Medicine, Sleep Disorders Centre and Neuromuscular Centre, University Hospital of Zurich, Zurich, Switzerland.,Lane Fox Respiratory Unit and Sleep Disorders Centre, Guy's and St Thomas' Hospital NHS Foundation Trust, London, United Kingdom
| | - Konrad E Bloch
- Department of Respiratory Medicine, Sleep Disorders Centre and Neuromuscular Centre, University Hospital of Zurich, Zurich, Switzerland,
| |
Collapse
|
41
|
Vijayvargiya P, Jameie-Oskooei S, Camilleri M, Chedid V, Erwin PJ, Murad MH. Association between delayed gastric emptying and upper gastrointestinal symptoms: a systematic review and meta-analysis. Gut 2019; 68:804-813. [PMID: 29860241 DOI: 10.1136/gutjnl-2018-316405] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 04/16/2018] [Accepted: 04/29/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND The relationship between delayed gastric emptying and upper GI symptoms (UGI Sx) is controversial. OBJECTIVE To assess association between gastric emptying and UGI Sx, independent of treatment. DESIGN We performed a systematic review and meta-analysis of the literature from 2007 to 2017, review of references and additional papers identified by content expert. We included studies evaluating the association between gastric emptying and nausea, vomiting, early satiety/postprandial fullness, abdominal pain and bloating. Covariate analyses included optimal gastric emptying test method, gastric emptying type (breath test or scintigraphy) and patient category. Meta-regression compared the differences based on type of gastric emptying tests. RESULTS Systematic review included 92 gastric emptying studies (26 breath test, 62 scintigraphy, 1 ultrasound and 3 wireless motility capsule); 25 of these studies provided quantitative data for meta-analysis (15 scintigraphy studies enrolling 4056 participants and 10 breath test studies enrolling 2231 participants). Meta-regression demonstrated a significant difference between optimal and suboptimal gastric emptying test methods when comparing delayed gastric emptying with nausea and vomiting. On evaluating studies using optimal gastric emptying test methodology, there were significant associations between gastric emptying and nausea (OR 1.6, 95% CI 1.4 to 1.8), vomiting (OR 2.0, 95% CI 1.6 to 2.7), abdominal pain (OR 1.5, 95% CI 1.0 to 2.2) and early satiety/fullness (OR 1.8, 95% CI 1.2 to 2.6) for patients with UGI Sx; gastric emptying and early satiety/fullness in patients with diabetes; gastric emptying and nausea in patients with gastroparesis. CONCLUSIONS The systematic review and meta-analysis supports an association between optimally measured delayed gastric emptying and UGI Sx.
Collapse
Affiliation(s)
- Priya Vijayvargiya
- Clinical Enteric Neuroscience Translational and Epidemiological Research (CENTER), Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Sina Jameie-Oskooei
- Clinical Enteric Neuroscience Translational and Epidemiological Research (CENTER), Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiological Research (CENTER), Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Victor Chedid
- Clinical Enteric Neuroscience Translational and Epidemiological Research (CENTER), Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Patricia J Erwin
- Division of Library Services, Mayo Clinic, Rochester, Minnesota, USA
| | | |
Collapse
|
42
|
Fiorentino G, Esquinas AM. Colonic distension treatment in Duchenne muscular dystrophy. Neuromuscul Disord 2019; 29:157-158. [PMID: 30744916 DOI: 10.1016/j.nmd.2018.10.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 10/27/2018] [Accepted: 10/29/2018] [Indexed: 11/16/2022]
Affiliation(s)
- Giuseppe Fiorentino
- Fisiopatologia e Riabilitazione Respiratoria, AO Ospedali dei Colli, Napoli PO, Monaldi, Italy.
| | - Antonio M Esquinas
- FCCP Internacional Fellow AARC, Intensive Care Unit, Hospital Morales Meseguer, Murcia, Spain
| |
Collapse
|
43
|
Mavroudis PD, van den Anker J, Conklin LS, Damsker JM, Hoffman EP, Nagaraju K, Clemens PR, Jusko WJ. Population Pharmacokinetics of Vamorolone (VBP15) in Healthy Men and Boys With Duchenne Muscular Dystrophy. J Clin Pharmacol 2019; 59:979-988. [PMID: 30742306 DOI: 10.1002/jcph.1388] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 01/18/2019] [Indexed: 02/06/2023]
Abstract
Duchenne muscular dystrophy (DMD) is an inherited neuromuscular disorder occurring in boys and caused by mutations in the dystrophin gene. Vamorolone is a first-generation delta-9,11 compound that has favorable efficacy and side effect profiles relative to classical glucocorticoids. The pharmacokinetics (PK) of oral vamorolone were assessed in parallel-group studies in healthy men (phase 1, n = 86) and boys with DMD (phase 2a, n = 48) during 14 days of once-daily dosing with a range of doses. Vamorolone exhibited moderate variability in PK, with the maximum plasma concentration usually occurring at 2-4 hours and a half-life of approximately 2 hours for all doses and days examined. Population PK modeling of all data together indicated that the PK of vamorolone can be well described by a 1-compartment model with zero-order absorption. Both men and boys showed a dose-linearity of PK parameters for the doses examined, with no accumulation of the drug during daily dosing. Ingestion with food resulted in markedly enhanced absorption of the drug, as tested in healthy men. There were similar PK of vamorolone in healthy men and DMD boys with apparent clearance averaging 2.0 L/h/kg in men and 1.7 L/h/kg in boys. Overall, vamorolone exhibited well-behaved linear PK, with similar profiles in healthy men and boys with DMD, moderate variability in PK parameters, and absorption and disposition profiles similar to those of classical glucocorticoids.
Collapse
Affiliation(s)
- Panteleimon D Mavroudis
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - John van den Anker
- ReveraGen Biopharma, Rockville, MD, USA.,Children's National Health System, Washington, DC, USA
| | - Laurie S Conklin
- ReveraGen Biopharma, Rockville, MD, USA.,Children's National Health System, Washington, DC, USA
| | | | - Eric P Hoffman
- ReveraGen Biopharma, Rockville, MD, USA.,Binghamton University-SUNY, Binghamton, NY, USA
| | - Kanneboyina Nagaraju
- ReveraGen Biopharma, Rockville, MD, USA.,Binghamton University-SUNY, Binghamton, NY, USA
| | - Paula R Clemens
- University of Pittsburgh and Department of Veterans Affairs Medical Center, Pittsburgh, PA, USA
| | - William J Jusko
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| |
Collapse
|
44
|
Metabolomics Research Reveals the Mechanism of Action of Astragalus Polysaccharide in Rats with Digestive System Disorders. Molecules 2018; 23:molecules23123333. [PMID: 30558291 PMCID: PMC6321338 DOI: 10.3390/molecules23123333] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 12/10/2018] [Accepted: 12/14/2018] [Indexed: 02/07/2023] Open
Abstract
With the diversity of modern dietary lifestyles, digestive system disorders (DSD) have become a frequently occurring disease in recent years. Astragalus polysaccharide (APS) is a homogeneous polysaccharide extracted from Astragalus, which might ameliorate the digestive and absorptive functions. However, the treatment mechanisms remain unclear. In this study, rats with DSD were fed a high-fat⁻low-protein diet and subjected to weight-bearing swimming until exhaustion. When body weight and autonomous activities of the rats decreased, they were administered APS. After two weeks, serum metabolomics analysis based on LC-MS was performed to validate the therapeutic effect of APS and explore its mechanism. APS pharmacodynamics was determined in this study, and serum metabolomics analysis discovered and identified 16 significant, differentially produced metabolites involved in energy, amino acid, and lipid metabolism, including citric acid, lactic acid, alanine, phosphatidylcholine, phenylalanine. After treatment with APS, the levels of the above small-molecule metabolites were reversed. Our results show the efficacy of APS in DSD treatment through the regulation of perturbed metabolic pathways related to energy, amino acid, and lipid metabolism.
Collapse
|
45
|
Nutrition in Duchenne muscular dystrophy 16–18 March 2018, Zaandam, the Netherlands. Neuromuscul Disord 2018; 28:680-689. [DOI: 10.1016/j.nmd.2018.05.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 05/09/2018] [Indexed: 11/17/2022]
|
46
|
Doris T, Bowron A, Armstrong A, Messer B. Ketoacidosis in Duchenne muscular dystrophy: A report on 4 cases. Neuromuscul Disord 2018; 28:665-670. [DOI: 10.1016/j.nmd.2018.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 06/05/2018] [Accepted: 06/08/2018] [Indexed: 10/14/2022]
|
47
|
Andrews JG, Wahl RA. Duchenne and Becker muscular dystrophy in adolescents: current perspectives. ADOLESCENT HEALTH MEDICINE AND THERAPEUTICS 2018; 9:53-63. [PMID: 29588625 PMCID: PMC5858539 DOI: 10.2147/ahmt.s125739] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD) are life-limiting and progressive neuromuscular conditions with significant comorbidities, many of which manifest during adolescence. BMD is a milder presentation of the condition and much less prevalent than DMD, making it less represented in the literature, or more severely affected individuals with BMD may be subsumed into the DMD population using clinical cutoffs. Numerous consensus documents have been published on the clinical management of DMD, the most recent of which was released in 2010. The advent of these clinical management consensus papers, particularly respiratory care, has significantly increased the life span for these individuals, and the adolescent years are now a point of transition into adult lives, rather than a period of end of life. This review outlines the literature on DMD and BMD during adolescence, focusing on clinical presentation during adolescence, impact of living with a chronic illness on adolescents, and the effect that adolescents have on their chronic illness. In addition, we describe the role that palliative-care specialists could have in improving outcomes for these individuals. The increasing proportion of individuals with DMD and BMD living into adulthood underscores the need for more research into interventions and intracacies of adolescence that can improve the social aspects of their lives.
Collapse
Affiliation(s)
| | - Richard A Wahl
- Department of Pediatrics, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
48
|
SchÄfer BT, Silveira MP, Palombit K, Mendes CE, Watanabe IS, Miglino MA, Castelucci P. Morphological Characterization of the Myenteric Plexus of the Ileum and Distal colon of Dogs Affected by Muscular Dystrophy. Anat Rec (Hoboken) 2017; 301:673-685. [DOI: 10.1002/ar.23708] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 02/02/2017] [Accepted: 05/03/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Bárbara Tavares SchÄfer
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science; University of São Paulo; Brazil
| | - Mariana Póvoa Silveira
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science; University of São Paulo; Brazil
| | - Kelly Palombit
- Department of Anatomy/Biomedical Sciences Institute; University of São Paulo; Brazil
- Deparment of Morphology; University Federal do Piaui; Teresina-PI Brazil
| | | | - Ii Sei Watanabe
- Department of Anatomy/Biomedical Sciences Institute; University of São Paulo; Brazil
| | - Maria Angélica Miglino
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science; University of São Paulo; Brazil
| | - Patricia Castelucci
- Department of Anatomy/Biomedical Sciences Institute; University of São Paulo; Brazil
| |
Collapse
|
49
|
Matsuo M, Awano H, Matsumoto M, Nagai M, Kawaguchi T, Zhang Z, Nishio H. Dystrophin Dp116: A yet to Be Investigated Product of the Duchenne Muscular Dystrophy Gene. Genes (Basel) 2017; 8:genes8100251. [PMID: 28974057 PMCID: PMC5664101 DOI: 10.3390/genes8100251] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 09/26/2017] [Indexed: 12/12/2022] Open
Abstract
The Duchenne muscular dystrophy (DMD) gene is one of the largest genes in the human genome. The gene exhibits a complex arrangement of seven alternative promoters, which drive the expression of three full length and four shorter isoforms. Dp116, the second smallest product of the DMD gene, is a Schwann cell-specific isoform encoded by a transcript corresponding to DMD exons 56–79, starting from a promoter/exon S1 within intron 55. The physiological roles of Dp116 are poorly understood, because of its extensive homology with other isoforms and its expression in specific tissues. This review summarizes studies on Dp116, focusing on clinical findings and alternative activation of the upstream translation initiation codon that is predicted to produce Dp118.
Collapse
Affiliation(s)
- Masafumi Matsuo
- Department of Physical Therapy, Faculty of Rehabilitation, Kobe Gakuin University, Kobe 651-2180, Japan.
| | - Hiroyuki Awano
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.
| | - Masaaki Matsumoto
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.
| | - Masashi Nagai
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.
| | - Tatsuya Kawaguchi
- Biomedical Analysis and Pathology Research Group, Discovery Science and Technology Department, Daiichi Sankyo RD Novare Co., Tokyo 134-8630, Japan.
| | - Zhujun Zhang
- Department of Physical Therapy, Faculty of Rehabilitation, Kobe Gakuin University, Kobe 651-2180, Japan.
| | - Hisahide Nishio
- Department of Community Medicine and Social Healthcare Sciences, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.
| |
Collapse
|