Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 132
Filter
Add more filters

Publication year range
1.
Mol Ther ; 32(9): 2930-2938, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-38850023

ABSTRACT

Lysosomal storage disorders (LSDs) are multisystemic progressive disorders caused by defects in proteins involved in lysosomal function. Different gene therapy strategies are under clinical investigation in several LSDs to overcome the limitations of available treatments. However, LSDs are slowly progressive diseases that require long-term studies to establish the efficacy of experimental treatments. Biomarkers can be reliable substitutes for clinical responses and improve the efficiency of clinical trials, especially when long-term disease interventions are evaluated. In this review, we summarize both available and future biomarkers for LSDs and discuss their strengths and weaknesses.


Subject(s)
Biomarkers , Clinical Trials as Topic , Genetic Therapy , Lysosomal Storage Diseases , Humans , Lysosomal Storage Diseases/therapy , Lysosomal Storage Diseases/genetics , Genetic Therapy/methods , Animals , Lysosomes/metabolism
2.
J Allergy Clin Immunol ; 153(3): 742-758, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38042501

ABSTRACT

BACKGROUND: Food allergy (FA) is one of the most common chronic conditions in children with an increasing prevalence facilitated by the exposure to environmental factors in predisposed individuals. It has been hypothesized that the increased consumption of ultra-processed foods, containing high levels of dietary advanced glycation end products (AGEs), could facilitate the occurrence of FA. OBJECTIVE: We sought to provide preclinical and clinical evidence on the potential role of AGEs in facilitating the occurrence of FA. METHODS: Human enterocytes, human small intestine organ culture, and PBMCs from children at risk for allergy were used to investigate the direct effect of AGEs on gut barrier, inflammation, TH2 cytokine response, and mitochondrial function. Intake of the 3 most common glycation products in Western diet foods, Nε-(carboxymethyl) lysine, Nε-(1-carboxyethyl) lysin, and Nδ-(5-hydro-5- methyl-4-imidazolone-2-yl)-ornithine (MG-H1), and the accumulation of AGEs in the skin were comparatively investigated in children with FA and in age-matched healthy controls. RESULTS: Human enterocytes exposed to AGEs showed alteration in gut barrier, AGE receptor expression, reactive oxygen species production, and autophagy, with increased transepithelial passage of food antigens. Small intestine organ cultures exposed to AGEs showed an increase of CD25+ cells and proliferating crypt enterocytes. PBMCs exposed to AGEs showed alteration in proliferation rate, AGE receptor activation, release of inflammatory and TH2 cytokines, and mitochondrial metabolism. Significant higher dietary AGE intake and skin accumulation were observed children with FA (n = 42) compared with age-matched healthy controls (n = 66). CONCLUSIONS: These data, supporting a potential role for dietary AGEs in facilitating the occurrence of FA, suggest the importance of limiting exposure to AGEs children as a potential preventive strategy against this common condition.


Subject(s)
Dietary Advanced Glycation End Products , Food Hypersensitivity , Child , Humans , Receptor for Advanced Glycation End Products , Glycation End Products, Advanced/metabolism , Diet, Western , Diet
3.
J Lipid Res ; : 100651, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39306041

ABSTRACT

Glycogen storage disease type Ia (GSDIa) is a rare, inherited glucose-6-phosphatase-α (G6Pase-α) deficiency-induced carbohydrate metabolism disorder. Although hyperlipidaemia is a hallmark of GSDI, the extent of lipid metabolism disruption remains incompletely understood. Lipidomic analysis was performed to characterise the serum lipidome in patients with GSDIa, by including age- and sex-matched healthy controls and age-matched hypercholesterolemic controls. Metabolic control and dietary information biochemical markers were obtained from patients with GSDIa. Patients with GSDIa showed higher total serum lysophosphatidylcholine (Fold Change, FC 2.2, p < 0.0001), acyl-acyl-phosphatidylcholine (FC 2.1, p < 0.0001), and ceramide (FC 2.4, p < 0.0001) levels and bile acid (FC 0.7, p < 0.001), acylcarnitines (FC 0.7, p < 0.001), and cholesterol esters (FC 1.0, p < 0.001) than those of healthy controls, and higher di- (FC 1.1, p < 0.0001; FC 0.9, p < 0.01) and triacylglycerol (FC 6.3, p < 0.0001; FC 3.9, p < 0.01) levels than those of healthy controls and hypercholesterolemic subjects. Both total cholesterol (TC) and TG values correlated with Cer(d16:1/22:0), Cer(d18:1/20:0), Cer(d18:1/20:0(OH)), Cer(d18:1/22:0), Cer(d18:1/23:0), Cer(d18:1/24:1), Cer(d18:2/22:0), Cer(d18:2/24:1). TC also correlated with Cer(d18:1/24:0), Cer(d18:2/20:0), HexCer(d16:1/22:0), HexCer(d18:1/18:0), and Hex2Cer(d18:1/20:0). TGlevels correlated with Cer(d18:0/24:1). Alanine transaminase values correlated with Cer(d18:0/22:0), insulin with Cer(d18:1/22:1) and Cer(d18:1/24:1), and HDL with hexosylceramide (HexCer)(d18:2/23:0). These results expand on the currently known involvement of lipid metabolism in GSDIa. Circulating Cer may allow for refined dietary assessment compared with traditional biomarkers. Because specific lipid species are relatively easy to assess, they represent potential novel biomarkers of GSDIa.

4.
Rev Endocr Metab Disord ; 25(4): 707-725, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38556561

ABSTRACT

Hepatic glycogen storage diseases constitute a group of disorders due to defects in the enzymes and transporters involved in glycogen breakdown and synthesis in the liver. Although hypoglycemia and hepatomegaly are the primary manifestations of (most of) hepatic GSDs, involvement of the endocrine system has been reported at multiple levels in individuals with hepatic GSDs. While some endocrine abnormalities (e.g., hypothalamic­pituitary axis dysfunction in GSD I) can be direct consequence of the genetic defect itself, others (e.g., osteopenia in GSD Ib, insulin-resistance in GSD I and GSD III) may be triggered by the (dietary/medical) treatment. Being aware of the endocrine abnormalities occurring in hepatic GSDs is essential (1) to provide optimized medical care to this group of individuals and (2) to drive research aiming at understanding the disease pathophysiology. In this review, a thorough description of the endocrine manifestations in individuals with hepatic GSDs is presented, including pathophysiological and clinical implications.


Subject(s)
Glycogen Storage Disease , Humans , Glycogen Storage Disease/metabolism , Glycogen Storage Disease/physiopathology , Liver Diseases/metabolism , Liver Diseases/physiopathology , Liver Diseases/etiology , Endocrine System Diseases/metabolism , Endocrine System Diseases/physiopathology , Liver/metabolism , Liver/physiopathology
5.
Eur J Neurol ; 31(9): e16383, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38873957

ABSTRACT

BACKGROUND AND PURPOSE: Two novel enzyme replacement therapies (ERTs), studied in phase 3 trials in late-onset Pompe patients, reached marketing authorization by the European Medicines Agency in 2022 and 2023. The European Pompe Consortium (EPOC) updates and extends the scope of the 2017 recommendations for starting, switching and stopping ERT. METHODS: The European Pompe Consortium consists of 25 neuromuscular and metabolic experts from eight European countries. This update was performed after an in-person meeting, three rounds of discussion and voting to provide a consensus recommendation. RESULTS: The patient should be symptomatic, that is, should have skeletal muscle weakness or respiratory muscle involvement. Muscle magnetic resonance imaging findings showing substantial fat replacement can support the decision to start in a patient-by-patient scenario. Limited evidence supports switching ERT if there is no indication that skeletal muscle and/or respiratory function have stabilized or improved during standard ERT of 12 months or after severe infusion-associated reactions. Switching of ERT should be discussed on a patient-by-patient shared-decision basis. If there are severe, unmanageable infusion-associated reactions and no stabilization in skeletal muscle function during the first 2 years after starting or switching treatment, stopping ERT should be considered. After stopping ERT for inefficacy, restarting ERT can be considered. Six-monthly European Pompe Consortium muscle function assessments are recommended. CONCLUSIONS: The triple-S criteria on ERT start, switch and stop include muscle magnetic resonance imaging as a supportive finding and the potential option of home infusion therapy. Six-monthly long-term monitoring of muscle function is highly recommended to cover insights into the patient's trajectory under ERT.


Subject(s)
Enzyme Replacement Therapy , Glycogen Storage Disease Type II , Glycogen Storage Disease Type II/drug therapy , Humans , Enzyme Replacement Therapy/methods , Europe
6.
J Enzyme Inhib Med Chem ; 36(1): 2068-2079, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34565280

ABSTRACT

Pompe disease is an inherited metabolic disorder due to the deficiency of the lysosomal acid α-glucosidase (GAA). The only approved treatment is enzyme replacement therapy with the recombinant enzyme (rhGAA). Further approaches like pharmacological chaperone therapy, based on the stabilising effect induced by small molecules on the target enzyme, could be a promising strategy. However, most known chaperones could be limited by their potential inhibitory effects on patient's enzymes. Here we report on the discovery of novel chaperones for rhGAA, L- and D-carnitine, and the related compound acetyl-D-carnitine. These drugs stabilise the enzyme at pH and temperature without inhibiting the activity and acted synergistically with active-site directed pharmacological chaperones. Remarkably, they enhanced by 4-fold the acid α-glucosidase activity in fibroblasts from three Pompe patients with added rhGAA. This synergistic effect of L-carnitine and rhGAA has the potential to be translated into improved therapeutic efficacy of ERT in Pompe disease.


Subject(s)
Carnitine/pharmacology , Glycoside Hydrolase Inhibitors/pharmacology , Lysosomes/drug effects , Molecular Chaperones/pharmacology , alpha-Glucosidases/metabolism , Allosteric Regulation/drug effects , Carnitine/chemistry , Dose-Response Relationship, Drug , Glycoside Hydrolase Inhibitors/chemistry , Humans , Lysosomes/enzymology , Molecular Chaperones/chemistry , Molecular Structure , Structure-Activity Relationship
7.
J Inherit Metab Dis ; 43(4): 770-777, 2020 07.
Article in English | MEDLINE | ID: mdl-32064649

ABSTRACT

A potential role of dietary lipids in the management of hepatic glycogen storage diseases (GSDs) has been proposed, but no consensus on management guidelines exists. The aim of this study was to describe current experiences with dietary lipid manipulations in hepatic GSD patients. An international study was set up to identify published and unpublished cases describing hepatic GSD patients with a dietary lipid manipulation. A literature search was performed according to the Cochrane Collaboration methodology through PubMed and EMBASE (up to December 2018). All delegates who attended the dietetics session at the IGSD2017, Groningen were invited to share unpublished cases. Due to multiple biases, only data on GSDIII were presented. A total of 28 cases with GSDIII and a dietary lipid manipulation were identified. Main indications were cardiomyopathy and/or myopathy. A high fat diet was the most common dietary lipid manipulation. A decline in creatine kinase concentrations (n = 19, P < .001) and a decrease in cardiac hypertrophy in paediatric GSDIIIa patients (n = 7, P < .01) were observed after the introduction with a high fat diet. This study presents an international cohort of GSDIII patients with different dietary lipid manipulations. High fat diet may be beneficial in paediatric GSDIIIa patients with cardiac hypertrophy, but careful long-term monitoring for potential complications is warranted, such as growth restriction, liver inflammation, and hepatocellular carcinoma development.


Subject(s)
Cardiomyopathies/etiology , Dietary Fats , Glycogen Storage Disease Type III/diet therapy , Cardiomyopathies/physiopathology , Child , Glycogen Storage Disease Type III/complications , Humans , Liver/pathology , Monitoring, Physiologic , Triglycerides/blood
8.
Int J Mol Sci ; 21(7)2020 Apr 04.
Article in English | MEDLINE | ID: mdl-32260444

ABSTRACT

The recent advancements in the knowledge of lysosomal biology and function have translated into an improved understanding of the pathophysiology of mucopolysaccharidoses (MPSs). The concept that MPS manifestations are direct consequences of lysosomal engorgement with undegraded glycosaminoglycans (GAGs) has been challenged by new information on the multiple biological roles of GAGs and by a new vision of the lysosome as a signaling hub involved in many critical cellular functions. MPS pathophysiology is now seen as the result of a complex cascade of secondary events that lead to dysfunction of several cellular processes and pathways, such as abnormal composition of membranes and its impact on vesicle fusion and trafficking; secondary storage of substrates; impairment of autophagy; impaired mitochondrial function and oxidative stress; dysregulation of signaling pathways. The characterization of this cascade of secondary cellular events is critical to better understand the pathophysiology of MPS clinical manifestations. In addition, some of these pathways may represent novel therapeutic targets and allow for the development of new therapies for these disorders.


Subject(s)
Glycosaminoglycans/metabolism , Mucopolysaccharidoses/pathology , Autophagy , Humans , Lysosomes/metabolism , Mucopolysaccharidoses/metabolism , Oxidative Stress , Protein Transport
9.
Genet Med ; 21(3): 591-600, 2019 03.
Article in English | MEDLINE | ID: mdl-29997386

ABSTRACT

PURPOSE: We studied microRNAs as potential biomarkers for Pompe disease. METHODS: We analyzed microRNA expression by small RNA-seq in tissues from the disease murine model at two different ages (3 and 9 months), and in plasma from Pompe patients. RESULTS: In the mouse model we found 211 microRNAs that were differentially expressed in gastrocnemii and 66 in heart, with a different pattern of expression at different ages. In a preliminary analysis in plasma from six patients 55 microRNAs were differentially expressed. Sixteen of these microRNAs were common to those dysregulated in mouse tissues. These microRNAs are known to modulate the expression of genes involved in relevant pathways for Pompe disease pathophysiology (autophagy, muscle regeneration, muscle atrophy). One of these microRNAs, miR-133a, was selected for further quantitative real-time polymerase chain reaction analysis in plasma samples from 52 patients, obtained from seven Italian and Dutch biobanks. miR-133a levels were significantly higher in Pompe disease patients than in controls and correlated with phenotype severity, with higher levels in infantile compared with late-onset patients. In three infantile patients miR-133a decreased after start of enzyme replacement therapy and evidence of clinical improvement. CONCLUSION: Circulating microRNAs may represent additional biomarkers of Pompe disease severity and of response to therapy.


Subject(s)
Glycogen Storage Disease Type II/diagnosis , Glycogen Storage Disease Type II/genetics , MicroRNAs/genetics , Adult , Animals , Biomarkers/blood , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Knockout , MicroRNAs/physiology , Middle Aged
10.
J Immunol ; 198(10): 3803-3808, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28389590

ABSTRACT

Glycogen storage disease type 1b (GSD-1b) is an autosomal-recessive disease caused by mutation of glucose-6-phosphate transporter and characterized by altered glycogen/glucose homeostasis. A higher frequency of autoimmune diseases has been observed in GSD-1b patients, but the molecular determinants leading to this phenomenon remain unknown. To address this question, we investigated the effect of glucose-6-phosphate transporter mutation on immune cell homeostasis and CD4+ T cell functions. In GSD-1b subjects, we found lymphopenia and a reduced capacity of T cells to engage glycolysis upon TCR stimulation. These phenomena associated with reduced expression of the FOXP3 transcription factor, lower suppressive function in peripheral CD4+CD25+FOXP3+ regulatory T cells, and an impaired capacity of CD4+CD25- conventional T cells to induce expression of FOXP3 after suboptimal TCR stimulation. These data unveil the metabolic determinant leading to an increased autoimmunity risk in GSD-1b patients.


Subject(s)
Autoimmunity , Glycogen Storage Disease Type I/immunology , Glycogen Storage Disease Type I/metabolism , Glycolysis , T-Lymphocytes, Regulatory/immunology , Adolescent , Adult , Antiporters/genetics , Antiporters/metabolism , CD4-Positive T-Lymphocytes/immunology , Child , Child, Preschool , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Glycogen Storage Disease Type I/physiopathology , Homeostasis , Humans , Infant , Lymphopenia/immunology , Lymphopenia/physiopathology , Male , Monosaccharide Transport Proteins/genetics , Monosaccharide Transport Proteins/metabolism , Mutation , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Young Adult
12.
Mol Genet Metab ; 123(3): 337-346, 2018 03.
Article in English | MEDLINE | ID: mdl-29397290

ABSTRACT

Multiple sulfatase deficiency (MSD) is an ultra-rare neurodegenerative disorder that results in defective sulfatase post-translational modification. Sulfatases in the body are activated by a unique protein, formylglycine-generating enzyme (FGE) that is encoded by SUMF1. When FGE is absent or insufficient, all 17 known human sulfatases are affected, including the enzymes associated with metachromatic leukodystrophy (MLD), several mucopolysaccharidoses (MPS II, IIIA, IIID, IVA, VI), chondrodysplasia punctata, and X-linked ichthyosis. As such, individuals demonstrate a complex and severe clinical phenotype that has not been fully characterized to date. In this report, we describe two individuals with distinct clinical presentations of MSD. Also, we detail a comprehensive systems-based approach to the management of individuals with MSD, from the initial diagnostic evaluation to unique multisystem issues and potential management options. As there have been no natural history studies to date, the recommendations within this report are based on published studies and consensus opinion and underscore the need for future research on evidence-based outcomes to improve management of children with MSD.


Subject(s)
Consensus , Multiple Sulfatase Deficiency Disease/therapy , Rare Diseases/therapy , Sulfatases/metabolism , Brain/diagnostic imaging , Brain/metabolism , Brain/pathology , Child, Preschool , Female , Humans , Male , Multiple Sulfatase Deficiency Disease/diagnosis , Multiple Sulfatase Deficiency Disease/etiology , Multiple Sulfatase Deficiency Disease/pathology , Mutation , Oxidoreductases Acting on Sulfur Group Donors , Protein Processing, Post-Translational/genetics , Rare Diseases/diagnosis , Rare Diseases/etiology , Sulfatases/deficiency
13.
BMC Med Genet ; 19(1): 183, 2018 10 11.
Article in English | MEDLINE | ID: mdl-30305043

ABSTRACT

BACKGROUND: Mucopolysaccharidosis-IVA (Morquio A disease) is a lysosomal disorder in which the abnormal accumulation of keratan sulfate and chondroitin-6-sulfate is consequent to mutations in the galactosamine-6-sulfatase (GALNS) gene. Since standard DNA sequencing analysis fails to detect about 16% of GALNS mutant alleles, gross DNA rearrangement screening and uniparental disomy evaluation are required to complete the molecular diagnosis. Despite this, the second pathogenic GALNS allele generally remains unidentified in ~ 5% of Morquio-A disease patients. METHODS: In an attempt to bridge the residual gap between clinical and molecular diagnosis, we performed an mRNA-based evaluation of three Morquio-A disease patients in whom the second mutant GALNS allele had not been identified. We also performed sequence analysis of the entire GALNS gene in two patients. RESULTS: Different aberrant GALNS mRNA transcripts were characterized in each patient. Analysis of these transcripts then allowed the identification, in one patient, of a disease-causing deep intronic GALNS mutation. The aberrant mRNA products identified in the other two individuals resulted in partial exon loss. Despite sequencing the entire GALNS gene region in these patients, the identity of a single underlying pathological lesion could not be unequivocally determined. We postulate that a combination of multiple variants, acting in cis, may synergise in terms of their impact on the splicing machinery. CONCLUSIONS: We have identified GALNS variants located within deep intronic regions that have the potential to impact splicing. These findings have prompted us to incorporate mRNA analysis into our diagnostic flow procedure for the molecular analysis of Morquio A disease.


Subject(s)
Chondroitinsulfatases/genetics , Mucopolysaccharidosis IV/genetics , Mutation , RNA Splicing , RNA, Messenger/genetics , Adolescent , Base Sequence , Chondroitinsulfatases/metabolism , DNA Mutational Analysis , Decision Trees , Exons , Female , Genotype , Humans , Introns , Male , Mucopolysaccharidosis IV/diagnosis , Mucopolysaccharidosis IV/metabolism , Mucopolysaccharidosis IV/physiopathology , RNA, Messenger/metabolism
14.
J Inherit Metab Dis ; 41(6): 985-995, 2018 11.
Article in English | MEDLINE | ID: mdl-29435782

ABSTRACT

BACKGROUND: Glycogen storage disease type I (GSDI) is an inborn error of carbohydrate metabolism caused by mutations of either the G6PC gene (GSDIa) or the SLC37A4 gene (GSDIb). GSDIa patients are at higher risk of developing insulin-resistance (IR). Mitochondrial dysfunction has been implicated in the development of IR. Mitochondrial dysfunction can demonstrate abnormalities in plama acylcarnitines (ACs) and urine organic acids (UOA). The aim of the study was to investigate the presence of mitochondrial impairment in GSDI patients and its possible connection with IR. METHODS: Fourteen GSDIa, seven GSDIb patients, 28 and 14 age and sex-matched controls, were enrolled. Plasma ACs, UOA, and surrogate markers of IR (HOMA-IR, QUICKI, ISI, VAI) were measured. RESULTS: GSDIa patients showed higher short-chain ACs and long-chain ACs levels and increased urinary excretion of lactate, pyruvate, 2-ketoglutarate, 3-methylglutaconate, adipate, suberate, aconitate, ethylmalonate, fumarate, malate, sebacate, 4-octenedioate, 3OH-suberate, and 3-methylglutarate than controls (p < 0.05). GSDIb patients showed higher C0 and C4 levels and increased urinary excretion of lactate, 3-methylglutarate and suberate than controls (p < 0.05). In GSDIa patients C18 levels correlated with insulin serum levels, HOMA-IR, QUICKI, and ISI; long-chain ACs levels correlated with cholesterol, triglycerides, ALT serum levels, and VAI. DISCUSSION: Increased plasma ACs and abnormal UOA profile suggest mitochondrial impairment in GSDIa. Correlation data suggest a possible connection between mitochondrial impairment and IR. We hypothesized that mitochondrial overload might generate by-products potentially affecting the insulin signaling pathway, leading to IR. On the basis of the available data, the possible pathomechanism for IR in GSDIa is proposed.


Subject(s)
Glycogen Storage Disease Type I/complications , Insulin Resistance , Mitochondrial Diseases/diagnosis , Mitochondrial Diseases/etiology , Acids/urine , Adolescent , Adult , Antiporters/genetics , Biomarkers/blood , Carnitine/analogs & derivatives , Carnitine/blood , Case-Control Studies , Child , Child, Preschool , Female , Glucose-6-Phosphatase/genetics , Humans , Insulin/blood , Linear Models , Male , Monosaccharide Transport Proteins/genetics , Multivariate Analysis , Urinalysis , Young Adult
16.
Annu Rev Med ; 66: 471-86, 2015.
Article in English | MEDLINE | ID: mdl-25587658

ABSTRACT

Lysosomal storage diseases are a group of rare, inborn, metabolic errors characterized by deficiencies in normal lysosomal function and by intralysosomal accumulation of undegraded substrates. The past 25 years have been characterized by remarkable progress in the treatment of these diseases and by the development of multiple therapeutic approaches. These approaches include strategies aimed at increasing the residual activity of a missing enzyme (enzyme replacement therapy, hematopoietic stem cell transplantation, pharmacological chaperone therapy and gene therapy) and approaches based on reducing the flux of substrates to lysosomes. As knowledge has improved about the pathophysiology of lysosomal storage diseases, novel targets for therapy have been identified, and innovative treatment approaches are being developed.


Subject(s)
Enzyme Replacement Therapy , Genetic Therapy , Hematopoietic Stem Cell Transplantation , Lysosomal Storage Diseases/therapy , Humans , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/metabolism , Molecular Chaperones
17.
Echocardiography ; 34(2): 240-249, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28070903

ABSTRACT

BACKGROUND: Mucopolysaccharidoses (MPS) are inherited lysosomal storage disorders caused by deficiency of required glycosaminoglycans breakdown enzymes, inducing cardiac involvement. Little is known about myocardial deformation involvement in MPS. Our aim was to assess biventricular structure and function in asymptomatic children with MPS using standard echo Doppler and 2D speckle tracking (STE). METHODS: Fifteen MPS children (one type I, six type II, three type III A, one III B, three IV A, one VI), asymptomatic for cardiac symptoms, and 15 age and sex-matched healthy controls underwent echo Doppler and STE. Left ventricular (LV) wall thicknesses, diameters, and mass were normalized by z-score. LV global longitudinal strain (GLS), global circumferential strain (GCS), global radial strain (GRS) at papillary muscles, LV twisting, and right ventricular (RV) GLS were measured. RESULTS: The two groups were comparable for body mass index, heart rate, and blood pressure. LV mass index and relative wall thickness were higher in MPS. Ejection fraction (EF), and s' velocity did not differ between the two groups. E/A ratio was lower and E/e' higher in MPS. Tricuspid annular plane systolic excursion, RV s' and e' were lower in MPS. LV GLS did not differ between the two groups, but GCS (P=.014), GRS (P=.023), twisting (P=.012), and RV GLS (P<.001) were lower in the MPS group. CONCLUSIONS: LV strain abnormalities are detectable in MPS pediatric patients, independently of MPS type, when EF is still normal. RV GLS is also involved consensually with TAPSE reduction. STE can be useful for detection of subclinical myocardial damage in MPS.


Subject(s)
Echocardiography , Mucopolysaccharidoses/complications , Ventricular Dysfunction/complications , Ventricular Dysfunction/diagnostic imaging , Child , Echocardiography, Doppler , Female , Heart Ventricles/diagnostic imaging , Heart Ventricles/physiopathology , Humans , Male , Mucopolysaccharidoses/physiopathology , Reproducibility of Results
18.
Mol Ther ; 23(7): 1138-1148, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25881001

ABSTRACT

Lysosomal storage disorders (LSDs) are a group of inborn metabolic diseases caused by mutations in genes that encode proteins involved in different lysosomal functions, in most instances acidic hydrolases. Different therapeutic approaches have been developed to treat these disorders. Pharmacological chaperone therapy (PCT) is an emerging approach based on small-molecule ligands that selectively bind and stabilize mutant enzymes, increase their cellular levels, and improve lysosomal trafficking and activity. Compared to other approaches, PCT shows advantages, particularly in terms of oral administration, broad biodistribution, and positive impact on patients' quality of life. After preclinical in vitro and in vivo studies, PCT is now being translated in the first clinical trials, either as monotherapy or in combination with enzyme replacement therapy, for some of the most prevalent LSDs. For some LSDs, the results of the first clinical trials are encouraging and warrant further development. Future research in the field of PCT will be directed toward the identification of novel chaperones, including new allosteric drugs, and the exploitation of synergies between chaperone treatment and other therapeutic approaches.


Subject(s)
Enzyme Stability/drug effects , Lysosomal Storage Diseases/therapy , Molecular Chaperones/therapeutic use , Humans , Ligands , Lysosomal Storage Diseases/genetics , Lysosomes/drug effects , Lysosomes/genetics , Lysosomes/pathology , Protein Biosynthesis/drug effects , Protein Folding/drug effects , Proteolysis/drug effects
20.
J Pediatr ; 166(4): 1079-82, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25641239

ABSTRACT

Angiotensin converting enzyme (ACE)-inhibitors decrease glomerular hyperfiltration but not microalbuminuria and proteinuria in glycogen storage disease type I. In the current study, we demonstrated that severe hyperlipidemia is associated with ACE-inhibitor ineffectiveness. We underline the importance of adequate metabolic control in glycogen storage disease type I. A combination therapy with ACE-inhibitors and lipid lowering drugs might be considered.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Forecasting , Glycogen Storage Disease Type I/complications , Hyperlipidemias/complications , Hypolipidemic Agents/therapeutic use , Kidney Diseases/etiology , Lipids/blood , Adolescent , Adult , Child , Disease Progression , Female , Follow-Up Studies , Glomerular Filtration Rate/drug effects , Glycogen Storage Disease Type I/blood , Glycogen Storage Disease Type I/drug therapy , Humans , Hyperlipidemias/blood , Hyperlipidemias/drug therapy , Incidence , Italy/epidemiology , Kidney Diseases/diagnosis , Kidney Diseases/epidemiology , Male , Prospective Studies , Retrospective Studies , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL