Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 272
Filter
1.
Orphanet J Rare Dis ; 19(1): 179, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38685110

ABSTRACT

Mucopolysaccharidoses (MPSs) are caused by a deficiency in the enzymes needed to degrade glycosaminoglycans (GAGs) in the lysosome. The storage of GAGs leads to the involvement of several systems and even to the death of the patient. In recent years, an increasing number of therapies have increased the treatment options available to patients. Early treatment is beneficial in improving the prognosis, but children with MPSs are often delayed in their diagnosis. Therefore, there is an urgent need to develop a method for early screening and diagnosis of the disease. Tandem mass spectrometry (MS/MS) is an analytical method that can detect multiple substrates or enzymes simultaneously. GAGs are reliable markers of MPSs. MS/MS can be used to screen children at an early stage of the disease, to improve prognosis by treating them before symptoms appear, to evaluate the effectiveness of treatment, and for metabolomic analysis or to find suitable biomarkers. In the future, MS/MS could be used to further identify suitable biomarkers for MPSs for early diagnosis and to detect efficacy.


Subject(s)
Mucopolysaccharidoses , Tandem Mass Spectrometry , Humans , Mucopolysaccharidoses/diagnosis , Mucopolysaccharidoses/metabolism , Tandem Mass Spectrometry/methods , Biomarkers/metabolism , Glycosaminoglycans/metabolism
2.
Cytokine ; 173: 156410, 2024 01.
Article in English | MEDLINE | ID: mdl-37924740

ABSTRACT

Recently, it has been shown disturbances in oxidant/antioxidant system and increases in some inflammatory markers in animal studies and in some Mucopolysaccharidoses (MPSs) patients. In this study, we aimed to determine the oxidative stress/antioxidant parameters and pro-inflammatory cytokine levels in the serum of MPS patients, in order to evaluate the possible role of inflammation in these patient groups regarding to accumulated metabolites. MPS I (n = 3), MPS II (n = 8), MPS III (n = 4), MPS IVA (n = 3), MPS VI (n = 3), and VII (n = 1) patients and 20 age-matched healthy subjects were included into the study. There was no statistically significant change in activities of SOD, Catalase, GSH-Px and lipid peroxidation levels in erythrocytes between the MPS patients and healthy controls. While IL-1alpha (p = 0.054), IL-6 (p = 0.008) levels, and chitotriosidase activity (p = 0.003) elevated in MPS3 patients, IL1α (p = 0.006), IL-1ß (p = 0.006), IL-6 (p = 0.006), IFNγ (p = 0.006), and NFκB (p = 0.006) levels increased in MPS-6 patients. Elevated levels of IL-6, IL1α and chitotriosidase activity demonstrated macrophage activation in MPSIII untreated with enzyme replacement. Our study showed for the first time that high levels of IL1α, IL-6, IL1ß and NFκB were present in MPSVI patients, demonstrating the induction of inflammation by dermatan sulphate. The low level of paraoxonase in MPSVI patients may be a good marker for cardiac involvement. Overall, this study provides important insights into the relationship between lysosomal storage of glycosaminoglycan and inflammation in MPS patients. It highlights possible pathways for the increased release of inflammatory molecules and suggests new targets for the development of treatments.


Subject(s)
Mucopolysaccharidoses , Mucopolysaccharidosis VI , Animals , Humans , Glycosaminoglycans/metabolism , Interleukin-6 , Antioxidants , Mucopolysaccharidoses/metabolism , Inflammation
3.
Mol Genet Metab ; 140(3): 107685, 2023 11.
Article in English | MEDLINE | ID: mdl-37604083

ABSTRACT

The mucopolysaccharidoses (MPS) are a family of inborn errors of metabolism resulting from a deficiency in a lysosomal hydrolase responsible for the degradation of glycosaminoglycans (GAG). From a biochemical standpoint, excessive urinary excretion of GAG has afforded first-tier laboratory investigations for diagnosis whereas newborn screening programs employ lysosomal hydrolase measurements. Given false positives are not uncommon, second-tier diagnostic testing relies on lysosomal hydrolase measurements following elevated urinary GAG, and newborn screening results are often corroborated with GAG determinations. Molecular genetics requires acknowledgement, as identifying pathogenic variants in the hydrolase genes confirms the diagnosis and allows cascade testing for families, but genetic variants of uncertain significance complicate this paradigm. Initiating cellular, tissue and organ damage that leads to an MPS phenotype is undoubtedly the accumulation of partially degraded GAG, and with mass spectrometry technologies now readily available in the biochemical genetics' laboratory, the ability to properly measure these GAG fragments has been realized. The most common approach involves bacterial lyase/hydrolase digestion of the long chain GAG polymers into their disaccharide units that can be measured by mass spectrometry. Another, less well-known method, the endogenous, non-reducing end method, does not require depolymerization of GAG but rather relies on the mass spectrometric measurement of the naturally produced oligosaccharides that arise from the enzyme deficiency. All MPS can be identified by this one method, and evidence to date shows it to be the only GAG analysis method that gives no false positives when employed as a first-tier laboratory diagnostic test and second-tier newborn screening test.


Subject(s)
Glycosaminoglycans , Mucopolysaccharidoses , Infant, Newborn , Humans , Glycosaminoglycans/metabolism , Neonatal Screening/methods , Tandem Mass Spectrometry/methods , Mucopolysaccharidoses/diagnosis , Mucopolysaccharidoses/genetics , Mucopolysaccharidoses/metabolism , Biomarkers , Hydrolases
4.
Biochem Biophys Res Commun ; 665: 107-117, 2023 07 12.
Article in English | MEDLINE | ID: mdl-37149983

ABSTRACT

Although mucopolysaccharidoses (MPS) are monogenic diseases, caused by mutations in genes coding for enzymes involved in degradation of glycosaminoglycans (GAGs), recent studies suggested that changes in expressions of various genes might cause secondary and tertiary cellular dysfunctions modulating the course of these diseases. In this report, we demonstrate that vesicle trafficking regulation is affected in fibroblasts derived from patients suffering from 11 different types of MPS due to changes in levels of crucial proteins (estimated by automated Western-blotting) involved in this process, including caveolin, clathrin, huntingtin (Htt), APPL1, EEA1, GOPC, Rab5, and Rab7. Microscopic studies confirmed these results, while investigations of tissue samples derived from the MPS I mouse model indicated differences between various organs in this matter. Moreover, transcriptomic analyses provided a global picture for changes in expressions of genes related to vesicle trafficking in MPS cells. We conclude that vesicle trafficking is dysregulated in MPS cells and changes in this process might contribute to the molecular mechanisms of this disease. Most probably, primary GAG storage might cause a cellular stress response leading to dysregulation of expression of many genes which, in turn, results in changes in cellular processes like vesicle trafficking. This can significantly modulate the course of the disease due to enhancing accumulation of GAGs and altering crucial cellular processes. This hypothesis has been supported by normalization of levels of clathrin in MPS cells treated with either an active form of the deficient GAG-degrading enzyme or a compound (5,7-dihydroxy-3-(4-hydroxyphenyl)-4H-1-benzopyran-4-one) indirectly reducing the efficiency of GAG synthesis.


Subject(s)
Mucopolysaccharidoses , Mice , Animals , Cell Line , Mucopolysaccharidoses/genetics , Mucopolysaccharidoses/drug therapy , Mucopolysaccharidoses/metabolism , Glycosaminoglycans/metabolism , Golgi Matrix Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism
5.
Biomolecules ; 13(3)2023 03 15.
Article in English | MEDLINE | ID: mdl-36979466

ABSTRACT

Impaired glycosaminoglycans (GAGs) catabolism may lead to a cluster of rare metabolic and genetic disorders called mucopolysaccharidoses (MPSs). Each subtype is caused by the deficiency of one of the lysosomal hydrolases normally degrading GAGs. Affected tissues accumulate undegraded GAGs in cell lysosomes and in the extracellular matrix, thus leading to the MPS complex clinical phenotype. Although each MPS may present with recognizable signs and symptoms, these may often overlap between subtypes, rendering the diagnosis difficult and delayed. Here, we performed an exploratory analysis to develop a model that predicts MPS subtypes based on UHPLC-MS/MS measurement of a urine free GAG profile (or GAGome). We analyzed the GAGome of 78 subjects (38 MPS, 37 healthy and 3 with other MPS symptom-overlapping disorders) using a standardized kit in a central-blinded laboratory. We observed several MPS subtype-specific GAGome changes. We developed a multivariable penalized Lasso logistic regression model that attained 91.2% balanced accuracy to distinguish MPS type II vs. III vs. any other subtype vs. not MPS, with sensitivity and specificity ranging from 73.3% to 91.7% and from 98.4% to 100%, depending on the predicted subtype. In conclusion, the urine GAGome was revealed to be useful in accurately discriminating the different MPS subtypes with a single UHPLC-MS/MS run and could serve as a reliable diagnostic test for a more rapid MPS biochemical diagnosis.


Subject(s)
Glycosaminoglycans , Mucopolysaccharidoses , Humans , Tandem Mass Spectrometry , Diagnosis, Differential , Mucopolysaccharidoses/diagnosis , Mucopolysaccharidoses/genetics , Mucopolysaccharidoses/metabolism , Hydrolases/genetics
6.
Genes (Basel) ; 14(2)2023 01 20.
Article in English | MEDLINE | ID: mdl-36833198

ABSTRACT

In this report, changes in the levels of various long non-coding RNAs (lncRNAs) were demonstrated for the first time in fibroblasts derived from patients suffering from 11 types/subtypes of mucopolysaccharidosis (MPS). Some kinds of lncRNA (SNHG5, LINC01705, LINC00856, CYTOR, MEG3, and GAS5) were present at especially elevated levels (an over six-fold change relative to the control cells) in several types of MPS. Some potential target genes for these lncRNAs were identified, and correlations between changed levels of specific lncRNAs and modulations in the abundance of mRNA transcripts of these genes (HNRNPC, FXR1, TP53, TARDBP, and MATR3) were found. Interestingly, the affected genes code for proteins involved in various regulatory processes, especially gene expression control through interactions with DNA or RNA regions. In conclusion, the results presented in this report suggest that changes in the levels of lncRNAs can considerably influence the pathomechanism of MPS through the dysregulation of the expression of certain genes, especially those involved in the control of the activities of other genes.


Subject(s)
Mucopolysaccharidoses , RNA, Long Noncoding , Humans , RNA, Long Noncoding/genetics , Fibroblasts/metabolism , Mucopolysaccharidoses/metabolism , RNA-Binding Proteins/metabolism , Nuclear Matrix-Associated Proteins/metabolism
7.
Exp Biol Med (Maywood) ; 247(18): 1639-1649, 2022 09.
Article in English | MEDLINE | ID: mdl-36000158

ABSTRACT

Mucopolysaccharidoses (MPS) are a group of lysosomal storage diseases caused by defects in genes coding for proteins involved in degradation of glycosaminoglycans (GAGs). These complex carbohydrates accumulate in cells causing their serious dysfunctions. Apart from the physical GAG storage, secondary and tertiary changes may contribute significantly to the pathomechanism of the disease. Among processes which were not systematically investigated in MPS cells to date there is the cell cycle. Here, we studied perturbances in this crucial cellular process in majority of MPS types. Transcriptomic analyses indicated that expression of many genes coding for proteins involved in the cell cycle is dysregulated in all tested MPS cells. Importantly, levels of transcripts of particular genes were changed in the same manner (i.e. either up- or down-regulated) in most or all types of the disease, indicating a common mechanism of the dysregulation. Flow cytometric studies demonstrated that the cell cycle is disturbed in all MPS types, with increased fractions of cells in the G0/G1 phase in most types and decreased fractions of cells in the G2/M phase in all types. We found that increased levels of cyclin D1 and disturbed timing of its appearance during the cell cycle may contribute to the mechanism of dysregulation of this process in MPS. Reduction of GAG levels by either a specific enzyme or genistein-mediated inhibition of synthesis of these compounds improved, but not fully corrected, the cell cycle in MPS fibroblasts. Therefore, it is suggested that combination of the therapeutic approaches devoted to reduction of GAG levels with cyclin D1 inhibitors might be considered in further works on developing effective treatment procedures for MPS.


Subject(s)
Genistein , Mucopolysaccharidoses , Humans , Genistein/pharmacology , Genistein/therapeutic use , Cyclin D1/metabolism , Transcriptome/genetics , Cell Line , Mucopolysaccharidoses/genetics , Mucopolysaccharidoses/metabolism , Mucopolysaccharidoses/therapy , Glycosaminoglycans , Cell Cycle/genetics , Cell Division
8.
Genes (Basel) ; 13(4)2022 03 26.
Article in English | MEDLINE | ID: mdl-35456399

ABSTRACT

Monogenic diseases are primarily caused by mutations in a single gene; thus, they are commonly recognized as genetic disorders with the simplest mechanisms. However, recent studies have indicated that the molecular mechanisms of monogenic diseases can be unexpectedly complicated, and their understanding requires complex studies at the molecular level. Previously, we have demonstrated that in mucopolysaccharidoses (MPS), a group of monogenic lysosomal storage diseases, several hundreds of genes reveal significant changes in the expression of various genes. Although the secondary effects of the primary biochemical defect and the inefficient degradation of glycosaminoglycans (GAGs) might be considered, the scale of the changes in the expression of a large fraction of genes cannot be explained by a block in one biochemical pathway. Here, we demonstrate that in cellular models of 11 types of MPS, the expression of genes coding for proteins involved in the regulation of the expression of many other genes at various stages (such as signal transduction, transcription, splicing, RNA degradation, translation, and others) is significantly disturbed relative to the control cells. This conclusion was based on transcriptomic studies, supported by biochemical analyses of levels of selected proteins encoded by genes revealing an especially high level of dysregulation in MPS (EXOSC9, SRSF10, RPL23, and NOTCH3 proteins were investigated). Interestingly, the reduction in GAGs levels, through the inhibition of their synthesis normalized the amounts of EXOSC9, RPL23, and NOTCH3 in some (but not all) MPS types, while the levels of SRSF10 could not be corrected in this way. These results indicate that different mechanisms are involved in the dysregulation of the expression of various genes in MPS, pointing to a potential explanation for the inability of some therapies (such as enzyme replacement therapy or substrate reduction therapy) to fully correct the physiology of MPS patients. We suggest that the disturbed expression of some genes, which appears as secondary or tertiary effects of GAG storage, might not be reversible, even after a reduction in the amounts of the storage material.


Subject(s)
Lysosomal Storage Diseases , Mucopolysaccharidoses , Cell Cycle Proteins/genetics , Gene Expression Regulation/genetics , Glycosaminoglycans/genetics , Glycosaminoglycans/metabolism , Humans , Mucopolysaccharidoses/genetics , Mucopolysaccharidoses/metabolism , Repressor Proteins/genetics , Serine-Arginine Splicing Factors/genetics , Transcriptome
9.
Metab Brain Dis ; 37(2): 299-310, 2022 02.
Article in English | MEDLINE | ID: mdl-34928474

ABSTRACT

Mucopolysaccharidoses (MPS) are a group of diseases caused by mutations resulting in deficiencies of lysosomal enzymes which lead to the accumulation of partially undegraded glycosaminoglycans (GAG). This phenomenon causes severe and chronic disturbances in the functioning of the organism, and leads to premature death. The metabolic defects affect also functions of the brain in most MPS types (except types IV, VI, and IX). The variety of symptoms, as well as the ineffectiveness of GAG-lowering therapies, question the early theory that GAG storage is the only cause of these diseases. As disorders of ion homeostasis increasingly turn out to be co-causes of the pathogenesis of various human diseases, the aim of this work was to determine the perturbations related to the maintenance of the ion balance at both the transcriptome and cellular levels in MPS. Transcriptomic studies, performed with fibroblasts derived from patients with all types/subtypes of MPS, showed extensive changes in the expression of genes involved in processes related to ion binding, transport and homeostasis. Detailed analysis of these data indicated specific changes in the expression of genes coding for proteins participating in the metabolism of Ca2+, Fe2+ and Zn2+. The results of tests carried out with the mouse MPS I model (Idua-/-) showed reductions in concentrations of these 3 ions in the liver and spleen. The results of these studies indicate for the first time ionic concentration disorders as possible factors influencing the course of MPS and show them as hypothetical, additional therapeutic targets for this rare disease.


Subject(s)
Mucopolysaccharidoses , Mucopolysaccharidosis I , Animals , Cell Line , Glycosaminoglycans/metabolism , Homeostasis , Humans , Mice , Mucopolysaccharidoses/metabolism , Transcriptome
10.
Int J Mol Sci ; 22(23)2021 Nov 23.
Article in English | MEDLINE | ID: mdl-34884458

ABSTRACT

The accumulation of glycosaminoglycans (GAGs) in bone and cartilage leads to progressive damage in cartilage that, in turn, reduces bone growth by the destruction of the growth plate, incomplete ossification, and growth imbalance. The mechanisms of pathophysiology related to bone metabolism in mucopolysaccharidoses (MPS) include impaired chondrocyte function and the failure of endochondral ossification, which leads to the release of inflammatory cytokines via the activation of Toll-like receptors by GAGs. Although improvements in the daily living of patients with MPS have been achieved with enzyme replacement, treatment for the bone disorder is limited. There is an increasing need to identify biomarkers related to bone and cartilage to evaluate the progressive status and to monitor the treatment of MPS. Recently, new analysis methods, such as proteomic analysis, have identified new biomarkers in MPS. This review summarizes advances in clinical bone metabolism and bone biomarkers.


Subject(s)
Biomarkers/metabolism , Bone and Bones/pathology , Mucopolysaccharidoses/metabolism , Bone Remodeling , Bone and Bones/metabolism , Cytokines/metabolism , Humans , Mucopolysaccharidoses/pathology , Proteomics
11.
Autophagy ; 17(11): 3875-3876, 2021 11.
Article in English | MEDLINE | ID: mdl-34407725

ABSTRACT

Mucopolysaccharidoses (MPS) are inherited metabolic diseases with strong neurological involvement. MPSs are caused by defects in lysosomal enzymes involved in the degradation of glycosaminoglycans (GAGs), which consequently accumulate into the lysosomes as primary storage. Macroautophagy/autophagy impairment is well known to drive neurodegeneration in MPSs, however, mechanisms underlying such dysfunction are still poorly understood. Recently, by studying a mouse model for MPS-III (Sanfilippo syndrome) we have shown that the progressive aggregation of amyloid proteins in neuronal cell bodies occurs downstream of the GAG storage and, in turn, impairs the autophagy pathway by affecting lysosomal-dependent autophagosome clearance.


Subject(s)
Autophagy , Mucopolysaccharidoses/metabolism , Protein Aggregation, Pathological/metabolism , Animals , Humans , Lysosomes/metabolism
12.
Int J Mol Sci ; 22(5)2021 Mar 09.
Article in English | MEDLINE | ID: mdl-33803318

ABSTRACT

Mucopolysaccharidoses (MPS) are inherited metabolic diseases characterized by accumulation of incompletely degraded glycosaminoglycans (GAGs) in lysosomes. Although primary causes of these diseases are mutations in genes coding for enzymes involved in lysosomal GAG degradation, it was demonstrated that storage of these complex carbohydrates provokes a cascade of secondary and tertiary changes affecting cellular functions. Potentially, this might lead to appearance of cellular disorders which could not be corrected even if the primary cause of the disease is removed. In this work, we studied changes in cellular organelles in MPS fibroblasts relative to control cells. All 11 types and subtypes of MPS were included into this study to obtain a complex picture of changes in organelles in this group of diseases. Two experimental approaches were employed, transcriptomic analyses and electron microscopic assessment of morphology of organelles. We analyzed levels of transcripts of genes grouped into two terms included into the QuickGO database, 'Cellular component organization' (GO:0016043) and 'Cellular anatomical entity' (GO:0110165), to find that number of transcripts with significantly changed levels in MPS fibroblasts vs. controls ranged from 109 to 322 (depending on MPS type) in GO:0016043, and from 70 to 208 in GO:0110165. This dysregulation of expression of genes crucial for proper structures and functions of various organelles was accompanied by severe changes in morphologies of lysosomes, nuclei, mitochondria, Golgi apparatus, and endoplasmic reticulum. Interestingly, some observed changes occurred in all/most MPS types while others were specific to particular disease types/subtypes. We suggest that severe changes in organelles in MPS cells might arise from dysregulation of expression of a battery of genes involved in organelles' structures and functions. Intriguingly, normalization of GAG levels by using recombinant human enzymes specific to different MPS types corrected morphologies of some, but not all, organelles, while it failed to improve regulation of expression of selected genes. These results might suggest reasons for inability of enzyme replacement therapy to correct all MPS symptoms, particularly if initiated at advanced stages of the disease.


Subject(s)
Fibroblasts , Gene Expression Regulation , Mucopolysaccharidoses , Organelles , Cell Line , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Humans , Mucopolysaccharidoses/metabolism , Mucopolysaccharidoses/pathology , Organelles/metabolism , Organelles/ultrastructure
13.
Carbohydr Polym ; 253: 117261, 2021 Feb 01.
Article in English | MEDLINE | ID: mdl-33278943

ABSTRACT

Mucopolysaccharidosis (MPS) are rare inherited diseases characterized by accumulation of lysosomal glycosaminoglycans, including heparan sulfate (HS). Patients exhibit progressive multi-visceral dysfunction and shortened lifespan mainly due to a severe cardiac/respiratory decline. Cathepsin V (CatV) is a potent elastolytic protease implicated in extracellular matrix (ECM) remodeling. Whether CatV is inactivated by HS in lungs from MPS patients remained unknown. Herein, CatV colocalized with HS in MPS bronchial epithelial cells. HS level correlated positively with the severity of respiratory symptoms and negatively to the overall endopeptidase activity of cysteine cathepsins. HS bound tightly to CatV and impaired its activity. Withdrawal of HS by glycosidases preserved exogenous CatV activity, while addition of Surfen, a HS antagonist, restored elastolytic CatV-like activity in MPS samples. Our data suggest that the pathophysiological accumulation of HS may be deleterious for CatV-mediated ECM remodeling and for lung tissue homeostasis, thus contributing to respiratory disorders associated to MPS diseases.


Subject(s)
Bronchi/metabolism , Cathepsins/metabolism , Cysteine Endopeptidases/metabolism , Epithelial Cells/metabolism , Heparitin Sulfate/metabolism , Mucopolysaccharidoses/metabolism , Severity of Illness Index , Adolescent , Animals , Bronchi/pathology , CHO Cells , Child , Child, Preschool , Cricetulus , Extracellular Matrix/metabolism , Female , Heparitin Sulfate/antagonists & inhibitors , Humans , Male , Mucopolysaccharidoses/pathology , Urea/analogs & derivatives , Urea/pharmacology , Young Adult
14.
Metab Brain Dis ; 35(8): 1353-1360, 2020 12.
Article in English | MEDLINE | ID: mdl-32886284

ABSTRACT

Recent studies indicated that apart from lysosomal storage of glycosaminoglycans (GAGs), secondary and tertiary changes in cellular processes may significantly contribute to development of disorders and symptoms occurring in mucopolysaccharidoses (MPS), a group of lysosomal storage diseases in which neurodegeneration is specific for most types and subtypes. In this report, using transcriptomic data, we demonstrate that regulation of hundreds of genes coding for proteins involved in regulations of various cellular processes is changed in cells derived from patients suffering from all types and subtypes of MPS. Among such genes there are 10 which expression is significantly changed in 9 or more (out of 11) MPS types/subtypes; they include IER3IP1, SAR1A, TMEM38B, PLCB4, SIN3B, ABHD5, SH3BP5, CAPG, PCOLCE2, and MN1. Moreover, there are several genes whose expression is changed over log2 > 4 times in some MPS types relative to control cells. The above analysis indicates that significant changes in expression of genes coding for various regulators of cellular processes may considerably contribute to development of cellular dysfunctions, and further appearance of specific symptoms of MPS, including neurodegeneration.


Subject(s)
Cellular Microenvironment/physiology , Fibroblasts/metabolism , Mucopolysaccharidoses/genetics , Mucopolysaccharidoses/metabolism , Transcriptome/physiology , Cell Line , Cells, Cultured , Fibroblasts/pathology , Gene Expression , Humans , Mucopolysaccharidoses/pathology
15.
Eur J Pharmacol ; 888: 173534, 2020 Dec 05.
Article in English | MEDLINE | ID: mdl-32877657

ABSTRACT

Mucopolysaccharidoses (MPS) represent a devastating group of lysosomal storage diseases (LSD) affecting approximately 1 in 25,000 individuals, where degradation of glycosaminoglycans (GAG) by lysosomal enzymes is impaired due to mutations causing defects in one of GAG-degrading enzymes. The most commonly used therapy for MPS is enzyme replacement therapy, consisting of application of an active form of the missing enzyme. However, supply of the missing enzyme is not enough in case of MPS types whose symptoms are expressed in central nervous system (CNS), as enzyme does not cross the blood-brain barrier. Moreover, even though enzyme replacement therapy for non-neuronopathic MPS IVA type is approved, it has a limited impact on bone abnormalities, that are one of main symptoms in the disease. Therefore, research into alternative therapeutic approaches for these types of MPS is highly desirable. One such alternative strategy is accelerated degradation of GAG by induction of autophagy. Autophagy is a process of lysosomal degradation of macromolecules that become abnormal or unnecessary for cells. One of the latest discoveries is that GAGs can also be such molecules. Potential drug should also cross blood-brain barrier and be safe in long-term therapy. It seems that one of the polyphenols, resveratrol, can meet the requirements. The mechanism of its action in autophagy stimulation is pleiotropic. Therefore, in this review, we will briefly discuss potential of resveratrol treatment for mucopolysaccharidosis through autophagy stimulation based on research in diseases with similar outcome.


Subject(s)
Antioxidants/therapeutic use , Autophagy/drug effects , Glycosaminoglycans/metabolism , Mucopolysaccharidoses/drug therapy , Mucopolysaccharidoses/metabolism , Resveratrol/therapeutic use , Animals , Antioxidants/pharmacology , Autophagy/physiology , Humans , Resveratrol/pharmacology
16.
Mol Genet Metab ; 131(1-2): 181-196, 2020.
Article in English | MEDLINE | ID: mdl-32917509

ABSTRACT

Neurological dysfunction represents a significant clinical component of many of the mucopolysaccharidoses (also known as MPS disorders). The accurate and consistent assessment of neuropsychological function is essential to gain a greater understanding of the precise natural history of these conditions and to design effective clinical trials to evaluate the impact of therapies on the brain. In 2017, an International MPS Consensus Panel published recommendations for best practice in the design and conduct of clinical studies investigating the effects of therapies on cognitive function and adaptive behavior in patients with neuronopathic mucopolysaccharidoses. Based on an International MPS Consensus Conference held in February 2020, this article provides updated consensus recommendations and expands the objectives to include approaches for assessing behavioral and social-emotional state, caregiver burden and quality of life in patients with all mucopolysaccharidoses.


Subject(s)
Brain/metabolism , Mucopolysaccharidoses/therapy , Nervous System Diseases/therapy , Physical Therapy Modalities , Brain/pathology , Clinical Trials as Topic , Cognitive Dysfunction/physiopathology , Humans , Mucopolysaccharidoses/genetics , Mucopolysaccharidoses/metabolism , Nervous System Diseases/genetics , Nervous System Diseases/metabolism , Problem Behavior , Quality of Life
17.
FEBS Lett ; 594(20): 3363-3370, 2020 10.
Article in English | MEDLINE | ID: mdl-32880920

ABSTRACT

We used transcriptomic (RNA-seq) analyses to determine whether patients suffering from all types and subtypes of mucopolysaccharidosis (MPS), a severe inherited metabolic disease, may be more susceptible to coronavirus disease 2019 (COVID-19). The expression levels of genes encoding proteins potentially involved in SARS-CoV-2 development were estimated in MPS cell lines. Four genes (GTF2F2, RAB18, TMEM97, PDE4DIP) coding for proteins potentially facilitating virus development were down-regulated, while two genes (FBN1, MFGE8), the products of which potentially interfere with virus propagation, were up-regulated in most MPS types. Although narrowing of respiratory tract and occurrence of thick mucus, characteristic of MPS, are risk factors for COVID-19, transcriptomic analyses suggest that MPS cells might be less, rather than more, susceptible to SARS-CoV-2 infection.


Subject(s)
COVID-19/genetics , Mucopolysaccharidoses/genetics , SARS-CoV-2/physiology , Virus Internalization , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/metabolism , COVID-19/metabolism , COVID-19/pathology , COVID-19/prevention & control , Cells, Cultured , Fibroblasts/metabolism , Fibroblasts/pathology , Fibroblasts/virology , Gene Expression Profiling , Genetic Predisposition to Disease , Humans , Mucopolysaccharidoses/metabolism , Mucopolysaccharidoses/pathology , Mucopolysaccharidoses/virology , SARS-CoV-2/pathogenicity , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Transcriptome
18.
Biochem J ; 477(17): 3433-3451, 2020 09 18.
Article in English | MEDLINE | ID: mdl-32856704

ABSTRACT

Mucopolysaccharidoses comprise a group of rare metabolic diseases, in which the lysosomal degradation of glycosaminoglycans (GAGs) is impaired due to genetically inherited defects of lysosomal enzymes involved in GAG catabolism. The resulting intralysosomal accumulation of GAG-derived metabolites consequently manifests in neurological symptoms and also peripheral abnormalities in various tissues like liver, kidney, spleen and bone. As each GAG consists of differently sulfated disaccharide units, it needs a specific, but also partly overlapping set of lysosomal enzymes to accomplish their complete degradation. Recently, we identified and characterized the lysosomal enzyme arylsulfatase K (Arsk) exhibiting glucuronate-2-sulfatase activity as needed for the degradation of heparan sulfate (HS), chondroitin sulfate (CS) and dermatan sulfate (DS). In the present study, we investigated the physiological relevance of Arsk by means of a constitutive Arsk knockout mouse model. A complete lack of glucuronate desulfation was demonstrated by a specific enzyme activity assay. Arsk-deficient mice show, in an organ-specific manner, a moderate accumulation of HS and CS metabolites characterized by 2-O-sulfated glucuronate moieties at their non-reducing ends. Pathophysiological studies reflect a rather mild phenotype including behavioral changes. Interestingly, no prominent lysosomal storage pathology like bone abnormalities were detected. Our results from the Arsk mouse model suggest a new although mild form of mucopolysacharidose (MPS), which we designate MPS type IIB.


Subject(s)
Arylsulfatases/metabolism , Chondroitin Sulfates/metabolism , Heparitin Sulfate/metabolism , Mucopolysaccharidoses/metabolism , Animals , Arylsulfatases/genetics , Chondroitin Sulfates/genetics , Enzyme Activation , Heparitin Sulfate/genetics , Mice , Mice, Knockout , Mucopolysaccharidoses/genetics
19.
Int J Mol Sci ; 21(9)2020 Apr 30.
Article in English | MEDLINE | ID: mdl-32366041

ABSTRACT

Although mucopolysaccharidoses (MPS), inherited metabolic diseases from the group of lysosomal storage diseases (LSD), are monogenic disorders, recent studies indicated that their molecular mechanisms are complicated. Storage of glycosaminoglycans (GAGs), arising from a deficiency in one of the enzymes involved in the degradation of these compounds, is the primary cause of each MPS type. However, dysfunctions of various cellular organelles and disturbance of cellular processes have been reported which contribute considerably to pathomechanisms of the disease. Here, we present a complex transcriptomic analysis in which all types and subtypes of MPS were investigated, with special emphasis on genes related to cell activation processes. Complex changes in expression of these genes were found in fibroblasts of all MPS types, with number of transcripts revealing higher or lower levels (relative to control fibroblasts) between 19 and over 50, depending on MPS type. Genes in which expression was significantly affected in most MPS types code for proteins involved in following processes, classified according to Gene Ontology knowledge database: cell activation, cell growth, cell recognition, and cell division. Levels of some transcripts (including CD9, CLU, MME and others) were especially significantly changed (over five times relative to controls). Our results are discussed in the light of molecular pathomechanisms of MPS, indicating that secondary and/or tertiary changes, relative to GAG storage, might significantly modulate cellular dysfunctions and contribute to molecular mechanisms of the disease. This may influence the efficacy of various therapies and suggests why various treatments are not fully effective in improving the complex symptoms of MPS.


Subject(s)
Lysosomal Storage Diseases/metabolism , Mucopolysaccharidoses/metabolism , Adolescent , Cell Cycle/genetics , Cell Cycle/physiology , Cell Proliferation/genetics , Cell Proliferation/physiology , Child , Child, Preschool , Female , Gene Ontology , Humans , Infant , Lysosomal Storage Diseases/genetics , Male , Transcriptome/genetics , Transcriptome/physiology
20.
Int J Mol Sci ; 21(8)2020 Apr 23.
Article in English | MEDLINE | ID: mdl-32340185

ABSTRACT

The aim of this review is to summarize the evidence on efficacy, effectiveness and safety of intravenous enzyme replacement therapy (ERT) available for mucopolysaccharidoses (MPSs) I, II, IVA, VI and VII, gained in phase III clinical trials and in observational post-approval studies. Post-marketing data are sometimes conflicting or controversial, possibly depending on disease severity, differently involved organs, age at starting treatment, and development of anti-drug antibodies (ADAs). There is general agreement that ERT is effective in reducing urinary glycosaminoglycans and liver and spleen volume, while heart and joints outcomes are variable in different studies. Effectiveness on cardiac valves, trachea and bronchi, hearing and eyes is definitely poor, probably due to limited penetration in the specific tissues. ERT does not cross the blood-brain barrier, with the consequence that the central nervous system is not cured by intravenously injected ERT. All patients develop ADAs but their role in ERT tolerance and effectiveness has not been well defined yet. Lack of reliable biomarkers contributes to the uncertainties about effectiveness. The data obtained from affected siblings strongly indicates the need of neonatal screening for treatable MPSs. Currently, other treatments are under evaluation and will surely help improve the prognosis of MPS patients.


Subject(s)
Enzyme Replacement Therapy , Mucopolysaccharidoses/drug therapy , Adolescent , Adult , Biomarkers , Child , Child, Preschool , Clinical Trials as Topic , Disease Management , Disease Susceptibility , Enzyme Replacement Therapy/adverse effects , Enzyme Replacement Therapy/methods , Humans , Infant , Male , Middle Aged , Mucopolysaccharidoses/diagnosis , Mucopolysaccharidoses/etiology , Mucopolysaccharidoses/metabolism , Symptom Assessment , Treatment Outcome , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...