1
|
Vargas-López V, Prada LF, Alméciga-Díaz CJ. Evidence of epigenetic landscape shifts in mucopolysaccharidosis IIIB and IVA. Sci Rep 2024; 14:3961. [PMID: 38368436 PMCID: PMC10874391 DOI: 10.1038/s41598-024-54626-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 02/14/2024] [Indexed: 02/19/2024] Open
Abstract
Lysosomal storage diseases (LSDs) are a group of monogenic diseases characterized by mutations in genes coding for proteins associated with the lysosomal function. Despite the monogenic nature, LSDs patients exhibit variable and heterogeneous clinical manifestations, prompting investigations into epigenetic factors underlying this phenotypic diversity. In this study, we focused on the potential role of epigenetic mechanisms in the pathogenesis of mucopolysaccharidosis IIIB (MPS IIIB) and mucopolysaccharidosis IVA (MPS IVA). We analyzed DNA methylation (5mC) and histone modifications (H3K14 acetylation and H3K9 trimethylation) in MPS IIIB and MPS IVA patients' fibroblasts and healthy controls. The findings revealed that global DNA hypomethylation is present in cell lines for both diseases. At the same time, histone acetylation was increased in MPS IIIB and MPS IVA cells in a donor-dependent way, further indicating a shift towards relaxed open chromatin in these MPS. Finally, the constitutive heterochromatin marker, histone H3K9 trimethylation, only showed reduced clustering in MPS IIIB cells, suggesting limited alterations in heterochromatin organization. These findings collectively emphasize the significance of epigenetic mechanisms in modulating the phenotypic variations observed in LSDs. While global DNA hypomethylation could contribute to the MPS pathogenesis, the study also highlights individual-specific epigenetic responses that might contribute to phenotypic heterogeneity. Further research into the specific genes and pathways affected by these epigenetic changes could provide insights into potential therapeutic interventions for these MPS and other LSDs.
Collapse
Affiliation(s)
- Viviana Vargas-López
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Cra. 7 No. 43-82 Edificio 54, Laboratorio 305A, Bogotá D.C., 110231, Colombia
| | - Luisa F Prada
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Cra. 7 No. 43-82 Edificio 54, Laboratorio 305A, Bogotá D.C., 110231, Colombia
| | - Carlos J Alméciga-Díaz
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Cra. 7 No. 43-82 Edificio 54, Laboratorio 305A, Bogotá D.C., 110231, Colombia.
| |
Collapse
|
2
|
Heinken A, El Kouche S, Guéant-Rodriguez RM, Guéant JL. Towards personalized genome-scale modeling of inborn errors of metabolism for systems medicine applications. Metabolism 2024; 150:155738. [PMID: 37981189 DOI: 10.1016/j.metabol.2023.155738] [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: 08/14/2023] [Revised: 11/09/2023] [Accepted: 11/12/2023] [Indexed: 11/21/2023]
Abstract
Inborn errors of metabolism (IEMs) are a group of more than 1000 inherited diseases that are individually rare but have a cumulative global prevalence of 50 per 100,000 births. Recently, it has been recognized that like common diseases, patients with rare diseases can greatly vary in the manifestation and severity of symptoms. Here, we review omics-driven approaches that enable an integrated, holistic view of metabolic phenotypes in IEM patients. We focus on applications of Constraint-based Reconstruction and Analysis (COBRA), a widely used mechanistic systems biology approach, to model the effects of inherited diseases. Moreover, we review evidence that the gut microbiome is also altered in rare diseases. Finally, we outline an approach using personalized metabolic models of IEM patients for the prediction of biomarkers and tailored therapeutic or dietary interventions. Such applications could pave the way towards personalized medicine not just for common, but also for rare diseases.
Collapse
Affiliation(s)
- Almut Heinken
- Inserm UMRS 1256 NGERE - Nutrition, Genetics, and Environmental Risk Exposure, University of Lorraine, Nancy F-54000, France.
| | - Sandra El Kouche
- Inserm UMRS 1256 NGERE - Nutrition, Genetics, and Environmental Risk Exposure, University of Lorraine, Nancy F-54000, France
| | - Rosa-Maria Guéant-Rodriguez
- Inserm UMRS 1256 NGERE - Nutrition, Genetics, and Environmental Risk Exposure, University of Lorraine, Nancy F-54000, France; National Center of Inborn Errors of Metabolism, University Regional Hospital Center of Nancy, Nancy F-54000, France
| | - Jean-Louis Guéant
- Inserm UMRS 1256 NGERE - Nutrition, Genetics, and Environmental Risk Exposure, University of Lorraine, Nancy F-54000, France; National Center of Inborn Errors of Metabolism, University Regional Hospital Center of Nancy, Nancy F-54000, France
| |
Collapse
|
3
|
Mucopolysaccharidoses: Cellular Consequences of Glycosaminoglycans Accumulation and Potential Targets. Int J Mol Sci 2022; 24:ijms24010477. [PMID: 36613919 PMCID: PMC9820209 DOI: 10.3390/ijms24010477] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/09/2022] [Accepted: 12/24/2022] [Indexed: 12/30/2022] Open
Abstract
Mucopolysaccharidoses (MPSs) constitute a heterogeneous group of lysosomal storage disorders characterized by the lysosomal accumulation of glycosaminoglycans (GAGs). Although lysosomal dysfunction is mainly affected, several cellular organelles such as mitochondria, endoplasmic reticulum, Golgi apparatus, and their related process are also impaired, leading to the activation of pathophysiological cascades. While supplying missing enzymes is the mainstream for the treatment of MPS, including enzyme replacement therapy (ERT), hematopoietic stem cell transplantation (HSCT), or gene therapy (GT), the use of modulators available to restore affected organelles for recovering cell homeostasis may be a simultaneous approach. This review summarizes the current knowledge about the cellular consequences of the lysosomal GAGs accumulation and discusses the use of potential modulators that can reestablish normal cell function beyond ERT-, HSCT-, or GT-based alternatives.
Collapse
|
4
|
Delivery and assessment of a CRISPR/nCas9-based genome editing system on in vitro models of mucopolysaccharidoses IVA assisted by magnetite-based nanoparticles. Sci Rep 2022; 12:15045. [PMID: 36057729 PMCID: PMC9440901 DOI: 10.1038/s41598-022-19407-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 08/29/2022] [Indexed: 12/27/2022] Open
Abstract
Mucopolysaccharidosis IV A (MPS IVA) is a lysosomal disorder caused by mutations in the GALNS gene. Consequently, the glycosaminoglycans (GAGs) keratan sulfate and chondroitin 6-sulfate accumulate in the lysosomal lumen. Although enzyme replacement therapy has shown essential advantages for the patients, several challenges remain to overcome, such as the limited impact on the bone lesion and recovery of oxidative profile. Recently, we validated a CRISPR/nCas9-based gene therapy with promising results in an in vitro MPS IVA model. In this study, we have expanded the use of this CRISPR/nCas9 system to several MPS IVA fibroblasts carrying different GALNS mutations. Considering the latent need to develop more safety vectors for gene therapy, we co-delivered the CRISPR/nCas9 system with a novel non-viral vector based on magnetoliposomes (MLPs). We found that the CRISPR/nCas9 treatment led to an increase in enzyme activity between 5 and 88% of wild-type levels, as well as a reduction in GAGs accumulation, lysosomal mass, and mitochondrial-dependent oxidative stress, in a mutation-dependent manner. Noteworthy, MLPs allowed to obtain similar results to those observed with the conventional transfection agent lipofectamine. Overall, these results confirmed the potential of CRISPR/nCas9 as a genome editing tool for treating MPS IVA. We also demonstrated the potential use of MLPs as a novel delivery system for CRISPR/nCas9-based therapies.
Collapse
|
5
|
Benincore-Flórez E, El-Azaz J, Solarte GA, Rodríguez A, Reyes LH, Alméciga-Díaz CJ, Cardona C. Iduronate-2-sulfatase interactome: Validation by Yeast Two-Hybrid Assay. Heliyon 2022; 8:e09031. [PMID: 35284671 PMCID: PMC8913312 DOI: 10.1016/j.heliyon.2022.e09031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/08/2021] [Accepted: 02/24/2022] [Indexed: 11/25/2022] Open
Abstract
Mucopolysaccharidosis type II (MPS II), also known as Hunter syndrome, is a rare X-linked recessive disease caused by a deficiency of the lysosomal enzyme iduronate-2-sulfatase (IDS), which activates intracellular accumulation of nonmetabolized glycosaminoglycans such as heparan sulfate and dermatan sulfate. This accumulation causes severe damage to several tissues, principally the central nervous system. Previously, we identified 187 IDS-protein interactions in the mouse brain. To validate a subset of these interactions, we selected and cloned the coding regions of 10 candidate genes to perform a targeted yeast two-hybrid assay. The results allowed the identification of the physical interaction of IDS with LSAMP and SYT1. Although the physiological relevance of these complexes is unknown, recent advances allow us to point out that these interactions could be involved in vesicular trafficking of IDS through the interaction with SYT1, as well as to the ability to form a transcytosis module between the cellular components of the blood-brain-barrier (BBB) through its interaction with LSAMP. These results may shed light on the role of IDS on cellular homeostasis and may also contribute to the understanding of MPS II physiopathology and the development of novel therapeutic strategies to transport recombinant IDS through the brain endothelial cells toward the brain parenchyma.
Collapse
|
6
|
Yaneske E, Zampieri G, Bertoldi L, Benvenuto G, Angione C. Genome-scale metabolic modelling of SARS-CoV-2 in cancer cells reveals an increased shift to glycolytic energy production. FEBS Lett 2021; 595:2350-2365. [PMID: 34409594 PMCID: PMC8427129 DOI: 10.1002/1873-3468.14180] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/02/2021] [Accepted: 08/15/2021] [Indexed: 01/08/2023]
Abstract
Cancer is considered a high‐risk condition for severe illness resulting from COVID‐19. The interaction between severe acute respiratory syndrome coronavirus‐2 (SARS‐CoV‐2) and human metabolism is key to elucidating the risk posed by COVID‐19 for cancer patients and identifying effective treatments, yet it is largely uncharacterised on a mechanistic level. We present a genome‐scale map of short‐term metabolic alterations triggered by SARS‐CoV‐2 infection of cancer cells. Through transcriptomic‐ and proteomic‐informed genome‐scale metabolic modelling, we characterise the role of RNA and fatty acid biosynthesis in conjunction with a rewiring in energy production pathways and enhanced cytokine secretion. These findings link together complementary aspects of viral invasion of cancer cells, while providing mechanistic insights that can inform the development of treatment strategies.
Collapse
Affiliation(s)
- Elisabeth Yaneske
- School of Computing, Engineering and Digital Technologies, Teesside University, Middlesbrough, UK
| | - Guido Zampieri
- School of Computing, Engineering and Digital Technologies, Teesside University, Middlesbrough, UK.,Department of Biology, University of Padua, Italy
| | | | | | - Claudio Angione
- School of Computing, Engineering and Digital Technologies, Teesside University, Middlesbrough, UK.,Healthcare Innovation Centre, Teesside University, Middlesbrough, UK.,Centre for Digital Innovation, Teesside University, Middlesbrough, UK
| |
Collapse
|
7
|
Echeverri-Peña OY, Salazar-Barreto DA, Rodríguez-Lopez A, González J, Alméciga-Díaz CJ, Verano-Guevara CH, Barrera LA. Use of a neuron-glia genome-scale metabolic reconstruction to model the metabolic consequences of the Arylsulphatase a deficiency through a systems biology approach. Heliyon 2021; 7:e07671. [PMID: 34381909 PMCID: PMC8340118 DOI: 10.1016/j.heliyon.2021.e07671] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 05/10/2021] [Accepted: 07/23/2021] [Indexed: 12/26/2022] Open
Abstract
Metachromatic leukodystrophy (MLD) is a human neurodegenerative disorder characterized by progressive damage on the myelin band in the nervous system. MLD is caused by the impaired function of the lysosomal enzyme Arylsulphatase A (ARSA). The physiopathology mechanisms and the biochemical consequences in the brain of ARSA deficiency are not entirely understood. In recent years, the use of genome-scale metabolic (GEM) models has been explored as a tool for the study of the biochemical alterations in MLD. Previously, we modeled the metabolic consequences of different lysosomal storage diseases using single GEMs. In the case of MLD, using a glia GEM, we previously predicted that the metabolism of glycosphingolipids and neurotransmitters was altered. The results also suggested that mitochondrial metabolism and amino acid transport were the main reactions affected. In this study, we extended the modeling of the metabolic consequences of ARSA deficiency through the integration of neuron and glial cell metabolic models. Cell-specific models were generated from Recon2, and these were used to create a neuron-glial bi-cellular model. We propose a workflow for the integration of this type of model and its subsequent study. The results predicted the impairment pathways involved in the transport of amino acids, lipids metabolism, and catabolism of purines and pyrimidines. The use of this neuron-glial GEM metabolic reconstruction allowed to improve the prediction capacity of the metabolic consequences of ARSA deficiency, which might pave the way for the modeling of the biochemical alterations of other inborn errors of metabolism with central nervous system involvement.
Collapse
Affiliation(s)
- Olga Y Echeverri-Peña
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - Diego A Salazar-Barreto
- Centro para la Optimización y Probabilidad Aplicada (COPA), Department of Industrial Engineering, Faculty of Engineering, Universidad de los Andes, Bogotá D.C., Colombia.,Grupo de Bioquímica Computacional, Estructural y Bioinformática, Department of Nutrition and Biochemistry, Faculty of Science, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Alexander Rodríguez-Lopez
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá D.C., Colombia.,Licenciatura en Química, Universidad Distrital Francisco Jose de Caldas, Bogota D.C., Colombia.,Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá D.C., Colombia
| | - Janneth González
- Grupo de Bioquímica Computacional, Estructural y Bioinformática, Department of Nutrition and Biochemistry, Faculty of Science, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Carlos J Alméciga-Díaz
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | | | - Luis A Barrera
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá D.C., Colombia.,Clínica de Errores Innatos del Metabolismo, Hospital Universitario San Ignacio, Bogotá D.C., Colombia
| |
Collapse
|
8
|
Puentes-Tellez MA, Lerma-Barbosa PA, Garzón-Jaramillo RG, Suarez DA, Espejo-Mojica AJ, Guevara JM, Echeverri OY, Solano-Galarza D, Uribe-Ardila A, Alméciga-Díaz CJ. A perspective on research, diagnosis, and management of lysosomal storage disorders in Colombia. Heliyon 2020; 6:e03635. [PMID: 32258481 PMCID: PMC7113438 DOI: 10.1016/j.heliyon.2020.e03635] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 02/21/2020] [Accepted: 03/18/2020] [Indexed: 11/29/2022] Open
Abstract
Lysosomal storage diseases (LSDs) are a group of about 50 inborn errors of metabolism characterized by the lysosomal accumulation of partially or non-degraded molecules due to mutations in proteins involved in the degradation of macromolecules, transport, lysosomal biogenesis or modulators of lysosomal environment. Significant advances have been achieved in the diagnosis, management, and treatment of LSDs patients. In terms of approved therapies, these include enzyme replacement therapy (ERT), substrate reduction therapy, hematopoietic stem cell transplantation, and pharmacological chaperone therapy. In this review, we summarize the Colombian experience in LSDs thorough the evidence published. We identified 113 articles published between 1995 and 2019 that included Colombian researchers or physicians, and which were mainly focused in Mucopolysaccharidoses, Pompe disease, Gaucher disease, Fabry disease, and Tay-Sachs and Sandhoff diseases. Most of these articles focused on basic research, clinical cases, and mutation reports. Noteworthy, implementation of the enzyme assay in dried blood samples, led to a 5-fold increase in the identification of LSD patients, suggesting that these disorders still remain undiagnosed in the country. We consider that the information presented in this review will contribute to the knowledge of a broad spectrum of LSDs in Colombia and will also contribute to the development of public policies and the identification of research opportunities.
Collapse
Affiliation(s)
- María Alejandra Puentes-Tellez
- Instituto de Errores Innatos del Metabolismo, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - Paula Andrea Lerma-Barbosa
- Instituto de Errores Innatos del Metabolismo, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | | | - Diego A. Suarez
- Instituto de Errores Innatos del Metabolismo, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
- Facultad de Medicina, Universidad Nacional de Colombia, Bogotá D.C., Colombia
| | - Angela J. Espejo-Mojica
- Instituto de Errores Innatos del Metabolismo, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - Johana M. Guevara
- Instituto de Errores Innatos del Metabolismo, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - Olga Yaneth Echeverri
- Instituto de Errores Innatos del Metabolismo, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - Daniela Solano-Galarza
- Instituto de Errores Innatos del Metabolismo, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - Alfredo Uribe-Ardila
- Research Center in Biochemistry, Universidad de los Andes, Bogotá D.C., Colombia
| | - Carlos J. Alméciga-Díaz
- Instituto de Errores Innatos del Metabolismo, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| |
Collapse
|
9
|
Design and applications of gene therapy vectors for mucopolysaccharidosis in Colombia. Gene Ther 2019; 27:104-107. [PMID: 31267008 DOI: 10.1038/s41434-019-0086-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 06/10/2019] [Indexed: 12/19/2022]
Abstract
The authors briefly describe their work in the construction of viral derived vectors for the use in gene therapy of muchopolysaccharide storage diseases (MPS), especially in Morquio A syndrome. The motivations to undertake that line of research about twenty years ago was the belief that gene therapy was the most plausible treatment for monogenic diseases due to the transient effect and its difficulty to reach bone tissue of the only effective treatment in use, the enzyme replacement therapy. The strategy used to increase the bone targeting was to include in the vectors an aspartic acid octapeptide that increases their affinity for the oppositely charged hydroxyapatite molecule of bone. It is also discussed the difficulties to do front line research in many developing countries, due to the extended belief that their research money should be mainly devoted to projects that render solutions in a very short time. However, the authors argue in favor of doing research in gene therapy, because it is proving to be the solution for many monogenic diseases, and therefore there is a need of people with good command of GT all over the world, in order to make good use of that therapy especially for ex-vivo treatments.
Collapse
|
10
|
Xu C, Qian Y. A selenamorpholine-based redox-responsive fluorescent probe for targeting lysosome and visualizing exogenous/endogenous hydrogen peroxide in living cells and zebrafish. J Mater Chem B 2019; 7:2714-2721. [PMID: 32255004 DOI: 10.1039/c8tb03010c] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
A simple selenamorpholine-based fluorescent probe has been designed and synthesized using a combination of selenamorpholine and a BODIPY fluorophore. BODIPY-Se has a low pKa value of 4.78 because of the selenamorpholine unit, which is beneficial for the probe to detect the lysosome. BODIPY-Se can turn on partial fluorescence only in lysosomes, due to a PET-inhibited process of protonation of selenamorpholine. In addition, the selenamorpholine unit of BODIPY-Se could selectively react with H2O2 through a redox reaction, leading to the alteration of the valence state of selenium from Se(ii) to Se(iv) and an additional PET-inhibited process. When BODIPY-Se tracked H2O2 in lysosomes, the two PET-inhibited processes would obviously amplify the fluorescence signal in living cells and in vivo. The probe could also detect the redox cycles between H2O2 and GSH continuously. Using confocal fluorescence imaging, the fluorescence localization of lysosomes demonstrated that BODIPY-Se could successfully target lysosomes. The probe could not only detect exogenous/endogenous H2O2 in living cells, but could also realize real-time monitoring of H2O2 in cancer cells and zebrafish. The results proved that BODIPY-Se is a promising fluorescent probe in biological applications.
Collapse
Affiliation(s)
- Chao Xu
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China.
| | | |
Collapse
|
11
|
Tebani A, Abily-Donval L, Schmitz-Afonso I, Piraud M, Ausseil J, Zerimech F, Pilon C, Pereira T, Marret S, Afonso C, Bekri S. Analysis of Mucopolysaccharidosis Type VI through Integrative Functional Metabolomics. Int J Mol Sci 2019; 20:ijms20020446. [PMID: 30669586 PMCID: PMC6359186 DOI: 10.3390/ijms20020446] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/17/2019] [Accepted: 01/18/2019] [Indexed: 12/12/2022] Open
Abstract
Metabolic phenotyping is poised as a powerful and promising tool for biomarker discovery in inherited metabolic diseases. However, few studies applied this approach to mcopolysaccharidoses (MPS). Thus, this innovative functional approach may unveil comprehensive impairments in MPS biology. This study explores mcopolysaccharidosis VI (MPS VI) or Maroteaux–Lamy syndrome (OMIM #253200) which is an autosomal recessive lysosomal storage disease caused by the deficiency of arylsulfatase B enzyme. Urine samples were collected from 16 MPS VI patients and 66 healthy control individuals. Untargeted metabolomics analysis was applied using ultra-high-performance liquid chromatography combined with ion mobility and high-resolution mass spectrometry. Furthermore, dermatan sulfate, amino acids, carnitine, and acylcarnitine profiles were quantified using liquid chromatography coupled to tandem mass spectrometry. Univariate analysis and multivariate data modeling were used for integrative analysis and discriminant metabolites selection. Pathway analysis was done to unveil impaired metabolism. The study revealed significant differential biochemical patterns using multivariate data modeling. Pathway analysis revealed that several major amino acid pathways were dysregulated in MPS VI. Integrative analysis of targeted and untargeted metabolomics data with in silico results yielded arginine-proline, histidine, and glutathione metabolism being the most affected. This study is one of the first metabolic phenotyping studies of MPS VI. The findings might shed light on molecular understanding of MPS pathophysiology to develop further MPS studies to enhance diagnosis and treatments of this rare condition.
Collapse
Affiliation(s)
- Abdellah Tebani
- Department of Metabolic Biochemistry, Rouen University Hospital, 76000 Rouen, France.
- Normandie University, UNIROUEN, CHU Rouen, INSERM U1245, 76000 Rouen, France.
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, COBRA, 76000 Rouen, France.
| | - Lenaig Abily-Donval
- Normandie University, UNIROUEN, CHU Rouen, INSERM U1245, 76000 Rouen, France.
- Department of Neonatal Pediatrics, Intensive Care and Neuropediatrics, Rouen University Hospital, 76031 Rouen, France.
| | | | - Monique Piraud
- Service de Biochimie et Biologie Moléculaire Grand Est, Unité des Maladies Héréditaires du Métabolisme et Dépistage Néonatal, Centre de Biologie et de Pathologie Est, Hospices Civils de Lyon, 69002 Lyon, France.
| | - Jérôme Ausseil
- INSERM U1088, Laboratoire de Biochimie Métabolique, Centre de Biologie Humaine, CHU Sud, 80054 Amiens CEDEX, France.
| | - Farid Zerimech
- Laboratoire de Biochimie et Biologie Moléculaire, Université de Lille et Pôle de Biologie Pathologie Génétique du CHRU de Lille, 59000 Lille, France.
| | - Carine Pilon
- Department of Metabolic Biochemistry, Rouen University Hospital, 76000 Rouen, France.
| | - Tony Pereira
- Department of Pharmacology, Rouen University Hospital, 76000 Rouen, France.
| | - Stéphane Marret
- Normandie University, UNIROUEN, CHU Rouen, INSERM U1245, 76000 Rouen, France.
- Department of Neonatal Pediatrics, Intensive Care and Neuropediatrics, Rouen University Hospital, 76031 Rouen, France.
| | - Carlos Afonso
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, COBRA, 76000 Rouen, France.
| | - Soumeya Bekri
- Department of Metabolic Biochemistry, Rouen University Hospital, 76000 Rouen, France.
- Normandie University, UNIROUEN, CHU Rouen, INSERM U1245, 76000 Rouen, France.
| |
Collapse
|
12
|
The induction of apoptosis in BEL-7402 cells by an iridium(III) complex through lysosome–mitochondria pathway. Polyhedron 2018. [DOI: 10.1016/j.poly.2018.09.057] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
13
|
Tebani A, Abily-Donval L, Schmitz-Afonso I, Héron B, Piraud M, Ausseil J, Zerimech F, Gonzalez B, Marret S, Afonso C, Bekri S. Unveiling metabolic remodeling in mucopolysaccharidosis type III through integrative metabolomics and pathway analysis. J Transl Med 2018; 16:248. [PMID: 30180851 PMCID: PMC6122730 DOI: 10.1186/s12967-018-1625-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 08/30/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Metabolomics represent a valuable tool to recover biological information using body fluids and may help to characterize pathophysiological mechanisms of the studied disease. This approach has not been widely used to explore inherited metabolic diseases. This study investigates mucopolysaccharidosis type III (MPS III). A thorough and holistic understanding of metabolic remodeling in MPS III may allow the development, improvement and personalization of patient care. METHODS We applied both targeted and untargeted metabolomics to urine samples obtained from a French cohort of 49 patients, consisting of 13 MPS IIIA, 16 MPS IIIB, 13 MPS IIIC, and 7 MPS IIID, along with 66 controls. The analytical strategy is based on ultra-high-performance liquid chromatography combined with ion mobility and high-resolution mass spectrometry. Twenty-four amino acids have been assessed using tandem mass spectrometry combined with liquid chromatography. Multivariate data modeling has been used for discriminant metabolite selection. Pathway analysis has been performed to retrieve metabolic pathways impairments. RESULTS Data analysis revealed distinct biochemical profiles. These metabolic patterns, particularly those related to the amino acid metabolisms, allowed the different studied groups to be distinguished. Pathway analysis unveiled major amino acid pathways impairments in MPS III mainly arginine-proline metabolism and urea cycle metabolism. CONCLUSION This represents one of the first metabolomics-based investigations of MPS III. These results may shed light on MPS III pathophysiology and could help to set more targeted studies to infer the biomarkers of the affected pathways, which is crucial for rare conditions such as MPS III.
Collapse
Affiliation(s)
- Abdellah Tebani
- Department of Metabolic Biochemistry, Rouen University Hospital, 76000, Rouen Cedex, France.,Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, 76000, Rouen, France.,Normandie Univ, UNIROUEN, INSA Rouen, CNRS, COBRA, 76000, Rouen, France
| | - Lenaig Abily-Donval
- Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, 76000, Rouen, France.,Department of Neonatal Pediatrics, Intensive Care and Neuropediatrics, Rouen University Hospital, 76031, Rouen, France
| | | | - Bénédicte Héron
- Department of Pediatric Neurology, Reference Center of Lysosomal Diseases, Trousseau Hospital, APHP and Sorbonne Université, GRC No 19, Pathologies Congénitales du Cervelet-LeucoDystrophies, AP-HP, Hôpital Armand Trousseau, 75012, Paris, France
| | - Monique Piraud
- Service de Biochimie et Biologie Moléculaire Grand Est, Unité des Maladies Héréditaires du Métabolisme et Dépistage Néonatal, Centre de Biologie et de Pathologie Est, CHU de Lyon, Lyon, France
| | - Jérôme Ausseil
- INSERM U1088, Laboratoire de Biochimie Métabolique, Centre de Biologie Humaine, CHU Sud, 80054, Amiens Cedex, France
| | - Farid Zerimech
- Laboratoire de Biochimie et Biologie Moléculaire, Université de Lille et Pôle de Biologie Pathologie Génétique du CHRU de Lille, 59000, Lille, France
| | - Bruno Gonzalez
- Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, 76000, Rouen, France
| | - Stéphane Marret
- Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, 76000, Rouen, France.,Department of Neonatal Pediatrics, Intensive Care and Neuropediatrics, Rouen University Hospital, 76031, Rouen, France
| | - Carlos Afonso
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, COBRA, 76000, Rouen, France
| | - Soumeya Bekri
- Department of Metabolic Biochemistry, Rouen University Hospital, 76000, Rouen Cedex, France. .,Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, 76000, Rouen, France.
| |
Collapse
|
14
|
Echeverri OY, Guevara JM, Espejo-Mojica ÁJ, Ardila A, Pulido N, Reyes M, Rodriguez-Lopez A, Alméciga-Díaz CJ, Barrera LA. Research, diagnosis and education in inborn errors of metabolism in Colombia: 20 years' experience from a reference center. Orphanet J Rare Dis 2018; 13:141. [PMID: 30115094 PMCID: PMC6097205 DOI: 10.1186/s13023-018-0879-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 07/26/2018] [Indexed: 12/11/2022] Open
Abstract
The use of specialized centers has been the main alternative for an appropriate diagnosis, management and follow up of patients affected by inborn errors of metabolism (IEM). These centers facilitate the training of different professionals, as well as the research at basic, translational and clinical levels. Nevertheless, few reports have described the experience of these centers and their local and/or global impact in the study of IEM. In this paper, we describe the experience of a Colombian reference center for the research, diagnosis, training and education on IEM. During the last 20 years, important advances have been achieved in the clinical knowledge of these disorders, as well as in the local availability of several diagnosis tests. Organic acidurias have been the most frequently detected diseases, followed by aminoacidopathies and peroxisomal disorders. Research efforts have been focused in the production of recombinant proteins in microorganisms towards the development of new enzyme replacement therapies, the design of gene therapy vectors and the use of bioinformatics tools for the understanding of IEM. In addition, this center has participated in the education and training of a large number professionals at different levels, which has contributed to increase the knowledge and divulgation of these disorders along the country. Noteworthy, in close collaboration with patient advocacy groups, we have participated in the discussion and construction of initiatives for the inclusion of diagnosis tests and treatments in the health system.
Collapse
Affiliation(s)
- Olga Y. Echeverri
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Cra. 7 No 43 - 82, Building 54, Room 305A, Bogotá, Colombia
| | - Johana M. Guevara
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Cra. 7 No 43 - 82, Building 54, Room 305A, Bogotá, Colombia
| | - Ángela J. Espejo-Mojica
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Cra. 7 No 43 - 82, Building 54, Room 305A, Bogotá, Colombia
| | - Andrea Ardila
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Cra. 7 No 43 - 82, Building 54, Room 305A, Bogotá, Colombia
- Clinical Laboratory – Inborn Errors of Metabolism Section, Hospital Universitario San Ignacio, Bogotá, Colombia
| | - Ninna Pulido
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Cra. 7 No 43 - 82, Building 54, Room 305A, Bogotá, Colombia
- Clinical Laboratory – Inborn Errors of Metabolism Section, Hospital Universitario San Ignacio, Bogotá, Colombia
| | - Magda Reyes
- Clinical Laboratory – Inborn Errors of Metabolism Section, Hospital Universitario San Ignacio, Bogotá, Colombia
| | - Alexander Rodriguez-Lopez
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Cra. 7 No 43 - 82, Building 54, Room 305A, Bogotá, Colombia
| | - Carlos J. Alméciga-Díaz
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Cra. 7 No 43 - 82, Building 54, Room 305A, Bogotá, Colombia
| | - Luis A. Barrera
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Cra. 7 No 43 - 82, Building 54, Room 305A, Bogotá, Colombia
- Clínica de Errores Innatos del Metabolismo, Hospital Universitario San Ignacio, Bogotá, Colombia
| |
Collapse
|
15
|
Fan J, Wang S, Zhang X, Chen W, Li Y, Yang P, Cao Z, Wang Y, Lu W, Ju D. Quantum Dots Elicit Hepatotoxicity through Lysosome-Dependent Autophagy Activation and Reactive Oxygen Species Production. ACS Biomater Sci Eng 2018; 4:1418-1427. [PMID: 33418671 DOI: 10.1021/acsbiomaterials.7b00824] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Jiajun Fan
- Minhang Branch, Zhongshan Hospital, Fudan University/Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201199, P. R. China
- Department of Microbiological and Biochemical Pharmacy and Key Lab of Smart Drug Delivery MOE, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| | - Shaofei Wang
- Minhang Branch, Zhongshan Hospital, Fudan University/Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201199, P. R. China
- Department of Microbiological and Biochemical Pharmacy and Key Lab of Smart Drug Delivery MOE, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| | - Xuyao Zhang
- Minhang Branch, Zhongshan Hospital, Fudan University/Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201199, P. R. China
- Department of Microbiological and Biochemical Pharmacy and Key Lab of Smart Drug Delivery MOE, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| | - Wei Chen
- Minhang Branch, Zhongshan Hospital, Fudan University/Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201199, P. R. China
- Department of Microbiological and Biochemical Pharmacy and Key Lab of Smart Drug Delivery MOE, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| | - Yubin Li
- Minhang Branch, Zhongshan Hospital, Fudan University/Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201199, P. R. China
- Department of Microbiological and Biochemical Pharmacy and Key Lab of Smart Drug Delivery MOE, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| | - Ping Yang
- Instrumental Analysis Center, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Zhonglian Cao
- Instrumental Analysis Center, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Yichen Wang
- Minhang Branch, Zhongshan Hospital, Fudan University/Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201199, P. R. China
- Department of Microbiological and Biochemical Pharmacy and Key Lab of Smart Drug Delivery MOE, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| | - Weiyue Lu
- Department of Microbiological and Biochemical Pharmacy and Key Lab of Smart Drug Delivery MOE, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| | - Dianwen Ju
- Minhang Branch, Zhongshan Hospital, Fudan University/Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201199, P. R. China
- Department of Microbiological and Biochemical Pharmacy and Key Lab of Smart Drug Delivery MOE, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| |
Collapse
|
16
|
Tebani A, Schmitz-Afonso I, Abily-Donval L, Héron B, Piraud M, Ausseil J, Brassier A, De Lonlay P, Zerimech F, Vaz FM, Gonzalez BJ, Marret S, Afonso C, Bekri S. Urinary metabolic phenotyping of mucopolysaccharidosis type I combining untargeted and targeted strategies with data modeling. Clin Chim Acta 2017; 475:7-14. [PMID: 28982054 DOI: 10.1016/j.cca.2017.09.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 09/29/2017] [Accepted: 09/30/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND Application of metabolic phenotyping could expand the pathophysiological knowledge of mucopolysaccharidoses (MPS) and may reveal the comprehensive metabolic impairments in MPS. However, few studies applied this approach to MPS. METHODS We applied targeted and untargeted metabolic profiling in urine samples obtained from a French cohort comprising 19 MPS I and 15 MPS I treated patients along with 66 controls. For that purpose, we used ultra-high-performance liquid chromatography combined with ion mobility and high-resolution mass spectrometry following a protocol designed for large-scale metabolomics studies regarding robustness and reproducibility. Furthermore, 24 amino acids have been quantified using liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS). Keratan sulfate, Heparan sulfate and Dermatan sulfate concentrations have also been measured using an LC-MS/MS method. Univariate and multivariate data analyses have been used to select discriminant metabolites. The mummichog algorithm has been used for pathway analysis. RESULTS The studied groups yielded distinct biochemical phenotypes using multivariate data analysis. Univariate statistics also revealed metabolites that differentiated the groups. Specifically, metabolites related to the amino acid metabolism. Pathway analysis revealed that several major amino acid pathways were dysregulated in MPS. Comparison of targeted and untargeted metabolomics data with in silico results yielded arginine, proline and glutathione metabolisms being the most affected. CONCLUSION This study is one of the first metabolic phenotyping studies of MPS I. The findings might help to generate new hypotheses about MPS pathophysiology and to develop further targeted studies of a smaller number of potentially key metabolites.
Collapse
Affiliation(s)
- Abdellah Tebani
- Department of Metabolic Biochemistry, Rouen University Hospital, Rouen 76000, France; Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, 76000 Rouen, France; Normandie Univ, UNIROUEN, INSA Rouen, CNRS, COBRA, 76000 Rouen, France
| | | | - Lenaig Abily-Donval
- Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, 76000 Rouen, France; Department of Neonatal Pediatrics and Intensive Care, Rouen University Hospital, Rouen 76031, France
| | - Bénédicte Héron
- Departement of Pediatric Neurology, Reference Center of Lysosomal Diseases, Trousseau Hospital, APHP, GRC ConCer-LD, Sorbonne Universities, UPMC University 06, Paris, France
| | - Monique Piraud
- Service de Biochimie et Biologie Moléculaire Grand Est, Unité des Maladies Héréditaires du Métabolisme et Dépistage Néonatal, Centre de Biologie et de Pathologie Est CHU de Lyon, Lyon, France
| | - Jérôme Ausseil
- INSERM U1088, Laboratoire de Biochimie Métabolique, Centre de Biologie Humaine, CHU Sud, 80054 Amiens Cedex, France
| | - Anais Brassier
- Reference Center of Inherited Metabolic Diseases, Imagine Institute, Hospital Necker Enfants Malades, APHP, University Paris Descartes, Paris, France
| | - Pascale De Lonlay
- Reference Center of Inherited Metabolic Diseases, Imagine Institute, Hospital Necker Enfants Malades, APHP, University Paris Descartes, Paris, France
| | - Farid Zerimech
- Laboratoire de Biochimie et Biologie Moléculaire, Université de Lille et Pôle de Biologie Pathologie Génétique du CHRU de Lille, 59000 Lille, France
| | - Frédéric M Vaz
- Laboratory of Genetic Metabolic Diseases, Department of Clinical Chemistry and Pediatrics, Academic Medical Center, Amsterdam, The Netherlands
| | - Bruno J Gonzalez
- Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, 76000 Rouen, France
| | - Stephane Marret
- Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, 76000 Rouen, France; Department of Neonatal Pediatrics and Intensive Care, Rouen University Hospital, Rouen 76031, France
| | - Carlos Afonso
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, COBRA, 76000 Rouen, France
| | - Soumeya Bekri
- Department of Metabolic Biochemistry, Rouen University Hospital, Rouen 76000, France; Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, 76000 Rouen, France.
| |
Collapse
|
17
|
Cvitanović T, Reichert MC, Moškon M, Mraz M, Lammert F, Rozman D. Large-scale computational models of liver metabolism: How far from the clinics? Hepatology 2017; 66:1323-1334. [PMID: 28520105 DOI: 10.1002/hep.29268] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 03/31/2017] [Accepted: 05/11/2017] [Indexed: 12/17/2022]
Abstract
Understanding the dynamics of human liver metabolism is fundamental for effective diagnosis and treatment of liver diseases. This knowledge can be obtained with systems biology/medicine approaches that account for the complexity of hepatic responses and their systemic consequences in other organs. Computational modeling can reveal hidden principles of the system by classification of individual components, analyzing their interactions and simulating the effects that are difficult to investigate experimentally. Herein, we review the state-of-the-art computational models that describe liver dynamics from metabolic, gene regulatory, and signal transduction perspectives. We focus especially on large-scale liver models described either by genome scale metabolic networks or an object-oriented approach. We also discuss the benefits and limitations of each modeling approach and their value for clinical applications in diagnosis, therapy, and prevention of liver diseases as well as precision medicine in hepatology. (Hepatology 2017;66:1323-1334).
Collapse
Affiliation(s)
- Tanja Cvitanović
- Centre for Functional Genomics and Bio-Chips, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Matthias C Reichert
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Miha Moškon
- Faculty of Computer and Information Science, University of Ljubljana, Ljubljana, Slovenia
| | - Miha Mraz
- Faculty of Computer and Information Science, University of Ljubljana, Ljubljana, Slovenia
| | - Frank Lammert
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Damjana Rozman
- Centre for Functional Genomics and Bio-Chips, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
18
|
Biomedical applications of cell- and tissue-specific metabolic network models. J Biomed Inform 2017; 68:35-49. [DOI: 10.1016/j.jbi.2017.02.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 02/21/2017] [Accepted: 02/23/2017] [Indexed: 12/17/2022]
|
19
|
Asghari A, Marashi SA, Ansari-Pour N. A sperm-specific proteome-scale metabolic network model identifies non-glycolytic genes for energy deficiency in asthenozoospermia. Syst Biol Reprod Med 2017; 63:100-112. [DOI: 10.1080/19396368.2016.1263367] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Arvand Asghari
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Sayed-Amir Marashi
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Naser Ansari-Pour
- Faculty of New Sciences and Technology, University of Tehran, Tehran, Iran
| |
Collapse
|
20
|
Khan S, Alméciga-Díaz CJ, Sawamoto K, Mackenzie WG, Theroux MC, Pizarro C, Mason RW, Orii T, Tomatsu S. Mucopolysaccharidosis IVA and glycosaminoglycans. Mol Genet Metab 2017; 120:78-95. [PMID: 27979613 PMCID: PMC5293636 DOI: 10.1016/j.ymgme.2016.11.007] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 11/16/2016] [Accepted: 11/16/2016] [Indexed: 12/21/2022]
Abstract
Mucopolysaccharidosis IVA (MPS IVA; Morquio A: OMIM 253000) is a lysosomal storage disease with an autosomal recessive trait caused by the deficiency of N-acetylgalactosamine-6-sulfate sulfatase. Deficiency of this enzyme leads to accumulation of specific glycosaminoglycans (GAGs): chondroitin-6-sulfate (C6S) and keratan sulfate (KS). C6S and KS are mainly produced in the cartilage. Therefore, the undegraded substrates are stored primarily in cartilage and in its extracellular matrix (ECM), leading to a direct impact on cartilage and bone development, and successive systemic skeletal dysplasia. Chondrogenesis, the earliest phase of skeletal formation, is maintained by cellular interactions with the ECM, growth and differentiation factors, signaling pathways, and transcription factors in a temporal-spatial manner. In patients with MPS IVA, the cartilage is disrupted at birth as a consequence of abnormal chondrogenesis and/or endochondral ossification. The unique skeletal features are distinguished by a disproportional short stature, odontoid hypoplasia, spinal cord compression, tracheal obstruction, pectus carinatum, kyphoscoliosis, platyspondyly, coxa valga, genu valgum, waddling gait, and laxity of joints. In spite of many descriptions of these unique clinical features, delay of diagnosis still happens. The pathogenesis and treatment of systemic skeletal dysplasia in MPS IVA remains an unmet challenge. In this review article, we comprehensively describe historical aspect, property of GAGs, diagnosis, screening, pathogenesis, and current and future therapies of MPS IVA.
Collapse
Affiliation(s)
- Shaukat Khan
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States
| | - Carlos J Alméciga-Díaz
- Institute for the Study of Inborn Errors of Metabolism, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - Kazuki Sawamoto
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States
| | - William G Mackenzie
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States
| | - Mary C Theroux
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States
| | - Christian Pizarro
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States
| | - Robert W Mason
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States
| | - Tadao Orii
- Department of Pediatrics, Gifu University, Gifu, Japan
| | - Shunji Tomatsu
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States; Department of Pediatrics, Gifu University, Gifu, Japan; Department of Pediatrics, Thomas Jefferson University, Philadelphia, PA, United States.
| |
Collapse
|
21
|
Hendriksz CJ. Elosulfase alfa (BMN 110) for the treatment of mucopolysaccharidosis IVA (Morquio A Syndrome). Expert Rev Clin Pharmacol 2016; 9:1521-1532. [PMID: 27855521 DOI: 10.1080/17512433.2017.1260000] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Morquio A syndrome is a rare, autosomal recessive, lysosomal storage disorder caused by a deficiency in the enzyme N-acetylgalactosamine-6-sulfatase (GALNS). In 2014, the use of recombinant human GALNS, elosulfase alfa, was approved in the European Union, Canada, the United States, Australia, and Brazil for the treatment of Morquio A syndrome. Elosulfase alfa is administered intravenously once-weekly at a dose of 2.0 mg/kg. Areas covered: This is a review of the efficacy, safety and tolerability, pharmacokinetics and pharmacodynamics, and other outcomes of elosulfase alfa treatment of patients with Morquio A. A discussion of other treatment considerations, limitations, and future directions in the use of elosulfase alfa is provided. Expert commentary: Pharmacokinetic studies outside of clinical trials and in 'real-world' clinical settings need to be performed. We cannot currently predict which patient is going to respond well to enzyme replacement therapy; thus, all patients should be given the option to receive treatment for at least 12 months. Additionally, accurate biomarkers for evaluating disease state and drug responsiveness would greatly aid in the treatment of patients with Morquio A. In addition, improved and innovative daily lifestyle measures are greatly needed to adequately measure clinical response and true impact on quality of life.
Collapse
Affiliation(s)
- Christian J Hendriksz
- a Paediatrics and Child Health , University of Pretoria , Steve Biko Academic Unit, Pretoria , South Africa
| |
Collapse
|