Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Genes (Basel) ; 15(3)2024 Feb 25.
Article in English | MEDLINE | ID: mdl-38540351

ABSTRACT

Rare diseases, or orphan diseases, are defined as diseases affecting a small number of people compared to the general population. Among these, we find lysosomal storage disorders (LSDs), a cluster of rare metabolic diseases characterized by enzyme mutations causing abnormal glycolipid storage. Drug repositioning involves repurposing existing approved drugs for new therapeutic applications, offering advantages in cost, time savings, and a lower risk of failure. We present a comprehensive analysis of existing drugs, their repurposing potential, and their clinical implications in the context of LSDs, highlighting the necessity of mutation-specific approaches. Our review systematically explores the landscape of drug repositioning as a means to enhance LSDs therapies. The findings advocate for the strategic repositioning of drugs, accentuating its role in expediting the discovery of effective treatments. We conclude that drug repurposing represents a viable pathway for accelerating therapeutic discovery for LSDs, emphasizing the need for the careful evaluation of drug efficacy and toxicity in disease-specific contexts.


Subject(s)
Drug Repositioning , Lysosomal Storage Diseases , Humans , Lysosomal Storage Diseases/drug therapy , Lysosomal Storage Diseases/genetics , Mutation , Lysosomes/metabolism
2.
Biochimie ; 222: 123-131, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38458414

ABSTRACT

PMM2-CDG, a disease caused by mutations in phosphomannomutase-2, is the most common congenital disorder of glycosylation. Yet, it still lacks a cure. Targeting phosphomannomutase-2 with pharmacological chaperones or inhibiting the phosphatase activity of phosphomannomutase-1 to enhance intracellular glucose-1,6-bisphosphate have been proposed as therapeutical approaches. We used Recombinant Bacterial Thermal Shift Assay to assess the binding of a substrate analog to phosphomannomutase-2 and the specific binding to phosphomannomutase-1 of an FDA-approved drug - clodronate. We also deepened the clodronate binding by enzyme activity assays and in silico docking. Our results confirmed the selective binding of clodronate to phosphomannomutase-1 and shed light on such binding.

3.
Biochim Biophys Acta Gen Subj ; 1868(2): 130526, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38049040

ABSTRACT

INTRODUCTION: The study of protein stability is crucial to biochemistry and relies on different methodologies. Recently, the Cellular Thermal Shift Assay has been introduced to study protein stability in whole cells. METHODS: We report a novel application of CeTSA named ReBaTSA. This Recombinant Bacterial TSA was performed using clear extracts from bacteria expressing a recombinant protein, incubated at different temperatures, centrifuged and analyzed via SDS-PAGE. RESULTS AND CONCLUSIONS: We demonstrated the feasibility and reliability of this simplified approach. We validated the method using the protein phosphomannomutase-2 and its common mutants, which were compared in the presence or the absence of a known ligand.


Subject(s)
Bacterial Lysates , Mutant Proteins , Reproducibility of Results , Protein Stability , Recombinant Proteins/genetics
4.
Orphanet J Rare Dis ; 18(1): 247, 2023 08 29.
Article in English | MEDLINE | ID: mdl-37644541

ABSTRACT

Congenital disorders of glycosylation are a group of more than 160 rare genetic defects in protein and lipid glycosylation. Since the first clinical report in 1980 of PMM2-CDG, the most common CDG worldwide, research made great strides, but nearly all of them are still missing a cure. CDG diagnosis has been at a rapid pace since the introduction of whole-exome/whole-genome sequencing as a diagnostic tool. Here, we retrace the history of CDG by analyzing all the patents associated with the topic. To this end, we explored the Espacenet database, extracted a list of patents, and then divided them into three major groups: (1) Drugs/therapeutic approaches for CDG, (2) Drug delivery tools for CDG, (3) Diagnostic tools for CDG. Despite the enormous scientific progress experienced in the last 30 years, diagnostic tools, drugs, and biomarkers are still urgently needed.


Subject(s)
Congenital Disorders of Glycosylation , Narration , Humans , Glycosylation , Congenital Disorders of Glycosylation/diagnosis , Congenital Disorders of Glycosylation/genetics , Databases, Factual , Exome
5.
Int J Mol Sci ; 24(5)2023 Feb 25.
Article in English | MEDLINE | ID: mdl-36901983

ABSTRACT

Enzyme replacement therapy is the only therapeutic option for Fabry patients with completely absent AGAL activity. However, the treatment has side effects, is costly, and requires conspicuous amounts of recombinant human protein (rh-AGAL). Thus, its optimization would benefit patients and welfare/health services (i.e., society at large). In this brief report, we describe preliminary results paving the way for two possible approaches: i. the combination of enzyme replacement therapy with pharmacological chaperones; and ii. the identification of AGAL interactors as possible therapeutic targets on which to act. We first showed that galactose, a low-affinity pharmacological chaperone, can prolong AGAL half-life in patient-derived cells treated with rh-AGAL. Then, we analyzed the interactomes of intracellular AGAL on patient-derived AGAL-defective fibroblasts treated with the two rh-AGALs approved for therapeutic purposes and compared the obtained interactomes to the one associated with endogenously produced AGAL (data available as PXD039168 on ProteomeXchange). Common interactors were aggregated and screened for sensitivity to known drugs. Such an interactor-drug list represents a starting point to deeply screen approved drugs and identify those that can affect (positively or negatively) enzyme replacement therapy.


Subject(s)
Fabry Disease , Humans , Fabry Disease/metabolism , alpha-Galactosidase/metabolism , Enzyme Replacement Therapy/methods , Isoenzymes/therapeutic use , Recombinant Proteins/therapeutic use
6.
Int J Mol Sci ; 24(2)2023 Jan 06.
Article in English | MEDLINE | ID: mdl-36674610

ABSTRACT

Fabry disease is a lysosomal storage disease caused by mutations in the GLA gene that encodes alpha-galactosidase (AGAL). The disease causes abnormal globotriaosylceramide (Gb3) storage in the lysosomes. Variants responsible for the genotypic spectrum of Fabry disease include mutations that abolish enzymatic activity and those that cause protein instability. The latter can be successfully treated with small molecules that either bind and stabilize AGAL or indirectly improve its cellular activity. This paper describes the first attempt to reposition curcumin, a nutraceutical, to treat Fabry disease. We tested the efficacy of curcumin in a cell model and found an improvement in AGAL activity for 80% of the tested mutant genotypes (four out of five tested). The fold-increase was dependent on the mutant and ranged from 1.4 to 2.2. We produced evidence that supports a co-chaperone role for curcumin when administered with AGAL pharmacological chaperones (1-deoxygalactonojirimycin and galactose). The combined treatment with curcumin and either pharmacological chaperone was beneficial for four out of five tested mutants and showed fold-increases ranging from 1.1 to 2.3 for DGJ and from 1.1 to 2.8 for galactose. Finally, we tested a long-term treatment on one mutant (L300F) and detected an improvement in Gb3 clearance and lysosomal markers (LAMP-1 and GAA). Altogether, our findings confirmed the necessity of personalized therapies for Fabry patients and paved the way to further studies and trials of treatments for Fabry disease.


Subject(s)
Curcumin , Fabry Disease , Humans , Fabry Disease/drug therapy , Fabry Disease/genetics , alpha-Galactosidase/metabolism , Curcumin/pharmacology , Curcumin/therapeutic use , Curcumin/metabolism , Galactose/metabolism , Mutation , Lysosomes/metabolism , 1-Deoxynojirimycin/pharmacology , 1-Deoxynojirimycin/therapeutic use
7.
Elife ; 112022 Oct 10.
Article in English | MEDLINE | ID: mdl-36214454

ABSTRACT

The most common cause of human congenital disorders of glycosylation (CDG) are mutations in the phosphomannomutase gene PMM2, which affect protein N-linked glycosylation. The yeast gene SEC53 encodes a homolog of human PMM2. We evolved 384 populations of yeast harboring one of two human-disease-associated alleles, sec53-V238M and sec53-F126L, or wild-type SEC53. We find that after 1000 generations, most populations compensate for the slow-growth phenotype associated with the sec53 human-disease-associated alleles. Through whole-genome sequencing we identify compensatory mutations, including known SEC53 genetic interactors. We observe an enrichment of compensatory mutations in other genes whose human homologs are associated with Type 1 CDG, including PGM1, which encodes the minor isoform of phosphoglucomutase in yeast. By genetic reconstruction, we show that evolved pgm1 mutations are dominant and allele-specific genetic interactors that restore both protein glycosylation and growth of yeast harboring the sec53-V238M allele. Finally, we characterize the enzymatic activity of purified Pgm1 mutant proteins. We find that reduction, but not elimination, of Pgm1 activity best compensates for the deleterious phenotypes associated with the sec53-V238M allele. Broadly, our results demonstrate the power of experimental evolution as a tool for identifying genes and pathways that compensate for human-disease-associated alleles.


Subject(s)
Congenital Disorders of Glycosylation , Saccharomyces cerevisiae Proteins , Humans , Saccharomyces cerevisiae/genetics , Congenital Disorders of Glycosylation/genetics , Congenital Disorders of Glycosylation/metabolism , Phosphoglucomutase/genetics , Mutant Proteins , Saccharomyces cerevisiae Proteins/genetics
8.
Int J Mol Sci ; 23(15)2022 Aug 05.
Article in English | MEDLINE | ID: mdl-35955863

ABSTRACT

Advances in research have boosted therapy development for congenital disorders of glycosylation (CDG), a group of rare genetic disorders affecting protein and lipid glycosylation and glycosylphosphatidylinositol anchor biosynthesis. The (re)use of known drugs for novel medical purposes, known as drug repositioning, is growing for both common and rare disorders. The latest innovation concerns the rational search for repositioned molecules which also benefits from artificial intelligence (AI). Compared to traditional methods, drug repositioning accelerates the overall drug discovery process while saving costs. This is particularly valuable for rare diseases. AI tools have proven their worth in diagnosis, in disease classification and characterization, and ultimately in therapy discovery in rare diseases. The availability of biomarkers and reliable disease models is critical for research and development of new drugs, especially for rare and heterogeneous diseases such as CDG. This work reviews the literature related to repositioned drugs for CDG, discovered by serendipity or through a systemic approach. Recent advances in biomarkers and disease models are also outlined as well as stakeholders' views on AI for therapy discovery in CDG.


Subject(s)
Congenital Disorders of Glycosylation , Artificial Intelligence , Biomarkers , Congenital Disorders of Glycosylation/genetics , Drug Repositioning , Humans , Rare Diseases
9.
Int J Mol Sci ; 23(9)2022 May 04.
Article in English | MEDLINE | ID: mdl-35563496

ABSTRACT

Fabry disease is caused by a deficiency of lysosomal alpha galactosidase and has a very large genotypic and phenotypic spectrum. Some patients who carry hypomorphic mutations can benefit from oral therapy with a pharmacological chaperone. The drug requires a very precise regimen because it is a reversible inhibitor of alpha-galactosidase. We looked for molecules that can potentiate this pharmacological chaperone, among drugs that have already been approved for other diseases. We tested candidate molecules in fibroblasts derived from a patient carrying a large deletion in the gene GLA, which were stably transfected with a plasmid expressing hypomorphic mutants. In our cell model, three drugs were able to potentiate the action of the pharmacological chaperone. We focused our attention on one of them, acetylsalicylic acid. We expect that acetylsalicylic acid can be used in synergy with the Fabry disease pharmacological chaperone and prolong its stabilizing effect on alpha-galactosidase.


Subject(s)
Fabry Disease , alpha-Galactosidase , 1-Deoxynojirimycin/pharmacology , 1-Deoxynojirimycin/therapeutic use , Aspirin/pharmacology , Aspirin/therapeutic use , Drug Repositioning , Fabry Disease/drug therapy , Fabry Disease/genetics , Humans , Lysosomes , Molecular Chaperones/genetics , Mutation , alpha-Galactosidase/genetics , alpha-Galactosidase/therapeutic use
10.
Genes (Basel) ; 12(4)2021 04 19.
Article in English | MEDLINE | ID: mdl-33921689

ABSTRACT

The protease encoded by the TMPRSS2 gene facilitates viral infections and has been implicated in the pathogenesis of SARS-CoV-2. We analyzed the TMPRSS2 sequence and correlated the protein variants with the clinical features of a cohort of 1177 patients affected by COVID-19 in Italy. Nine relatively common variants (allele frequency > 0.01) and six missense variants which may affect the protease activity according to PolyPhen-2 in HumVar-trained mode were identified. Among them, p.V197M (p.Val197Met) (rs12329760) emerges as a common variant that has a deleterious effect on the protease and a protective effect on the patients. Its role appears particularly relevant in two subgroups of patients-young males and elderly women-and among those affected by co-morbidities, where the variant frequency is higher among individuals who were mildly affected by the disease and did not need hospitalization or oxygen therapy than among those more severely affected, who required oxygen therapy, ventilation or intubation. This study provides useful information for the identification of patients at risk of developing a severe form of COVID-19, and encourages the usage of drugs affecting the expression of TMPRSS2 or inhibiting protein activity.


Subject(s)
COVID-19/etiology , Polymorphism, Single Nucleotide , Serine Endopeptidases/genetics , Aged , COVID-19/epidemiology , COVID-19/genetics , COVID-19/therapy , Comorbidity , Female , Gene Frequency , Hospitalization , Humans , Italy/epidemiology , Male , Middle Aged , Mutation , Respiration, Artificial , Serine Endopeptidases/chemistry , Serine Endopeptidases/metabolism , Treatment Outcome
11.
Eur J Med Genet ; 64(6): 104227, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33872774

ABSTRACT

The identification of high-risk factors for the infection by SARS-CoV-2 and the negative outcome of COVID-19 is crucial. The genetic background of the host might account for individual responses to SARS-CoV-2 infection besides age and comorbidities. A list of candidate polymorphisms is needed to drive targeted screens, given the existence of frequent polymorphisms in the general population. We carried out text mining in the scientific literature to draw up a list of genes referable to the term "SARS-CoV*". We looked for frequent mutations that are likely to affect protein function in these genes. Ten genes, mostly involved in innate immunity, and thirteen common variants were identified, for some of these the involvement in COVID-19 is supported by publicly available epidemiological data. We looked for available data on the population distribution of these variants and we demonstrated that the prevalence of five of them, Arg52Cys (rs5030737), Gly54Asp (rs1800450) and Gly57Glu (rs1800451) in MBL2, Ala59Thr (rs25680) in CD27, and Val197Met (rs12329760) in TMPRSS2, correlates with the number of cases and/or deaths of COVID-19 observed in different countries. The association of the TMPRSS2 variant provides epidemiological evidence of the usefulness of transmembrane protease serine 2 inhibitors for the cure of COVID-19. The identified genetic variants represent a basis for the design of a cost-effective assay for population screening of genetic risk factors in the COVID-19 pandemic.


Subject(s)
COVID-19/genetics , COVID-19/immunology , Genetic Predisposition to Disease , Immunity, Innate , SARS-CoV-2/pathogenicity , Data Mining , Gene Frequency , Genetic Variation , Host Microbial Interactions , Humans , Mannose-Binding Lectin/genetics , Mannose-Binding Lectin/immunology , Polymorphism, Single Nucleotide , Risk Factors , Serine Endopeptidases/genetics , Serine Endopeptidases/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology
12.
Molecules ; 25(9)2020 May 08.
Article in English | MEDLINE | ID: mdl-32397334

ABSTRACT

The 3,3',5,5'-tetrachloro-2-iodo-4,4'-bipyridine structure is proposed as a novel chemical scaffold for the design of new transthyretin (TTR) fibrillogenesis inhibitors. In the frame of a proof-of-principle exploration, four chiral 3,3',5,5'-tetrachloro-2-iodo-2'-substituted-4,4'- bipyridines were rationally designed and prepared from a simple trihalopyridine in three steps, including a Cu-catalysed Finkelstein reaction to introduce iodine atoms on the heteroaromatic scaffold, and a Pd-catalysed coupling reaction to install the 2'-substituent. The corresponding racemates, along with other five chiral 4,4'-bipyridines containing halogens as substituents, were enantioseparated by high-performance liquid chromatography in order to obtain pure enantiomer pairs. All stereoisomers were tested against the amyloid fibril formation (FF) of wild type (WT)-TTR and two mutant variants, V30M and Y78F, in acid mediated aggregation experiments. Among the 4,4'-bipyridine derivatives, interesting inhibition activity was obtained for both enantiomers of the 3,3',5,5'-tetrachloro-2'-(4-hydroxyphenyl)-2-iodo-4,4'-bipyridine. In silico docking studies were carried out in order to explore possible binding modes of the 4,4'-bipyridine derivatives into the TTR. The gained results point out the importance of the right combination of H-bond sites and the presence of iodine as halogen-bond donor. Both experimental and theoretical evidences pave the way for the utilization of the iodinated 4,4'-bipyridine core as template to design new promising inhibitors of TTR amyloidogenesis.


Subject(s)
Amyloid/chemistry , Hydrocarbons, Iodinated , Molecular Docking Simulation , Prealbumin/chemistry , Protein Aggregates , Pyridines , Amino Acid Substitution , Amyloid/genetics , Humans , Hydrocarbons, Iodinated/chemical synthesis , Hydrocarbons, Iodinated/chemistry , Mutation, Missense , Prealbumin/genetics , Pyridines/chemical synthesis , Pyridines/chemistry
13.
Biochem J ; 477(2): 359-380, 2020 01 31.
Article in English | MEDLINE | ID: mdl-31899485

ABSTRACT

The lysosomal storage disorder Fabry disease is characterized by a deficiency of the lysosomal enzyme α-Galactosidase A. The observation that missense variants in the encoding GLA gene often lead to structural destabilization, endoplasmic reticulum retention and proteasomal degradation of the misfolded, but otherwise catalytically functional enzyme has resulted in the exploration of alternative therapeutic approaches. In this context, we have investigated proteostasis regulators (PRs) for their potential to increase cellular enzyme activity, and to reduce the disease-specific accumulation of the biomarker globotriaosylsphingosine in patient-derived cell culture. The PRs also acted synergistically with the clinically approved 1-deoxygalactonojirimycine, demonstrating the potential of combination treatment in a therapeutic application. Extensive characterization of the effective PRs revealed inhibition of the proteasome and elevation of GLA gene expression as paramount effects. Further analysis of transcriptional patterns of the PRs exposed a variety of genes involved in proteostasis as potential modulators. We propose that addressing proteostasis is an effective approach to discover new therapeutic targets for diseases involving folding and trafficking-deficient protein mutants.


Subject(s)
Fabry Disease/genetics , Lysosomal Storage Diseases/genetics , Proteostasis/genetics , alpha-Galactosidase/genetics , 1-Deoxynojirimycin/analogs & derivatives , 1-Deoxynojirimycin/therapeutic use , Biomarkers/metabolism , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Fabry Disease/drug therapy , Fabry Disease/enzymology , Fabry Disease/pathology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Humans , Lysosomal Storage Diseases/drug therapy , Lysosomal Storage Diseases/enzymology , Lysosomal Storage Diseases/pathology , Lysosomes/enzymology , Lysosomes/genetics , Lysosomes/metabolism , Mutation, Missense/genetics , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Transport/drug effects , Sphingosine/analogs & derivatives , Sphingosine/metabolism
14.
Int J Mol Sci ; 21(2)2020 Jan 13.
Article in English | MEDLINE | ID: mdl-31940970

ABSTRACT

The term "pharmacological chaperone" was introduced 20 years ago. Since then the approach with this type of drug has been proposed for several diseases, lysosomal storage disorders representing the most popular targets. The hallmark of a pharmacological chaperone is its ability to bind a protein specifically and stabilize it. This property can be beneficial for curing diseases that are associated with protein mutants that are intrinsically active but unstable. The total activity of the affected proteins in the cell is lower than normal because they are cleared by the quality control system. Although most pharmacological chaperones are reversible competitive inhibitors or antagonists of their target proteins, the inhibitory activity is neither required nor desirable. This issue is well documented by specific examples among which those concerning Fabry disease. Direct specific binding is not the only mechanism by which small molecules can rescue mutant proteins in the cell. These drugs and the properly defined pharmacological chaperones can work together with different and possibly synergistic modes of action to revert a disease phenotype caused by an unstable protein.


Subject(s)
Fabry Disease , Molecular Chaperones/therapeutic use , Mutation, Missense , alpha-Galactosidase , Fabry Disease/drug therapy , Fabry Disease/enzymology , Fabry Disease/genetics , Humans , alpha-Galactosidase/genetics , alpha-Galactosidase/metabolism
15.
Int J Mol Sci ; 20(17)2019 Aug 26.
Article in English | MEDLINE | ID: mdl-31454904

ABSTRACT

A large number of mutations causing PMM2-CDG, which is the most frequent disorder of glycosylation, destabilize phosphomannomutase2. We looked for a pharmacological chaperone to cure PMM2-CDG, starting from the structure of a natural ligand of phosphomannomutase2, α-glucose-1,6-bisphosphate. The compound, ß-glucose-1,6-bisphosphate, was synthesized and characterized via 31P-NMR. ß-glucose-1,6-bisphosphate binds its target enzyme in silico. The binding induces a large conformational change that was predicted by the program PELE and validated in vitro by limited proteolysis. The ability of the compound to stabilize wild type phosphomannomutase2, as well as frequently encountered pathogenic mutants, was measured using thermal shift assay. ß-glucose-1,6-bisphosphate is relatively resistant to the enzyme that specifically hydrolyses natural esose-bisphosphates.


Subject(s)
Congenital Disorders of Glycosylation/genetics , Congenital Disorders of Glycosylation/metabolism , Glucose-6-Phosphate/analogs & derivatives , Mutation , Phosphotransferases (Phosphomutases)/deficiency , Congenital Disorders of Glycosylation/drug therapy , Glucose-6-Phosphate/chemistry , Glucose-6-Phosphate/metabolism , Glucose-6-Phosphate/pharmacology , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Conformation , Phosphotransferases (Phosphomutases)/genetics , Protein Binding
16.
Data Brief ; 22: 903-908, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30723759

ABSTRACT

The data presented in this article are connected to our research article entitled "D2A-Ala peptide derived from the urokinase receptor exerts anti-tumoural effects in vitro and in vivo" (Furlan et al., 2018). These data further extend our understanding of the inhibitory effects of D2A-Ala peptide. Dose-response curve using a wide range of concentrations of D2A-Ala shows that this peptide has no effects per se on proliferation of rat smooth muscle cells (RSMC). However, D2A-Ala dose-dependently inhibits epidermal growth factor (EGF)-induced RSMC proliferation. Kinetics lasting up to seven days revealed that D2A-Ala peptide completely blocked EGF-promoted RSMC proliferation. Moreover, D2A-Ala peptide inhibited invasion of HT 1080 cells towards RSMC.

17.
BMC Bioinformatics ; 19(Suppl 15): 433, 2018 Nov 30.
Article in English | MEDLINE | ID: mdl-30497360

ABSTRACT

BACKGROUND: Severity gradation of missense mutations is a big challenge for exome annotation. Predictors of deleteriousness that are most frequently used to filter variants found by next generation sequencing, produce qualitative predictions, but also numerical scores. It has never been tested if these scores correlate with disease severity. RESULTS: wANNOVAR, a popular tool that can generate several different types of deleteriousness-prediction scores, was tested on Fabry disease. This pathology, which is caused by a deficit of lysosomal alpha-galactosidase, has a very large genotypic and phenotypic spectrum and offers the possibility of associating a quantitative measure of the damage caused by mutations to the functioning of the enzyme in the cells. Some predictors, and in particular VEST3 and PolyPhen2 provide scores that correlate with the severity of lysosomal alpha-galactosidase mutations in a statistically significant way. CONCLUSIONS: Sorting disease mutations by severity is possible and offers advantages over binary classification. Dataset for testing and training in silico predictors can be obtained by transient transfection and evaluation of residual activity of mutants in cell extracts. This approach consents to quantitative data for severe, mild and non pathological variants.


Subject(s)
Lysosomes/enzymology , Molecular Sequence Annotation/methods , Mutation, Missense/genetics , alpha-Galactosidase/genetics , Fabry Disease/enzymology , Fabry Disease/genetics , Humans , Phenotype
18.
Int J Mol Sci ; 19(8)2018 Jul 30.
Article in English | MEDLINE | ID: mdl-30061496

ABSTRACT

Type I disorders of glycosylation (CDG), the most frequent of which is phosphomannomutase 2 (PMM2-CDG), are a group of diseases causing the incomplete N-glycosylation of proteins. PMM2-CDG is an autosomal recessive disease with a large phenotypic spectrum, and is associated with mutations in the PMM2 gene. The biochemical analysis of mutants does not allow a precise genotype⁻phenotype correlation for PMM2-CDG. PMM2 is very tolerant to missense and loss of function mutations, suggesting that a partial deficiency of activity might be beneficial under certain circumstances. The patient phenotype might be influenced by variants in other genes associated with the type I disorders of glycosylation in the general population.


Subject(s)
Congenital Disorders of Glycosylation/diagnosis , Congenital Disorders of Glycosylation/genetics , Mutation , Phosphotransferases (Phosphomutases)/genetics , Genetic Association Studies , Glycosylation , Humans , Models, Molecular , Mutation, Missense , Phenotype , Phosphotransferases (Phosphomutases)/chemistry , Protein Conformation
19.
Int J Mol Sci ; 19(5)2018 Apr 27.
Article in English | MEDLINE | ID: mdl-29702557

ABSTRACT

Congenital disorders of glycosylation (CDG) are a group of genetic disorders that affect protein and lipid glycosylation and glycosylphosphatidylinositol synthesis. More than 100 different disorders have been reported and the number is rapidly increasing. Since glycosylation is an essential post-translational process, patients present a large range of symptoms and variable phenotypes, from very mild to extremely severe. Only for few CDG, potentially curative therapies are being used, including dietary supplementation (e.g., galactose for PGM1-CDG, fucose for SLC35C1-CDG, Mn2+ for TMEM165-CDG or mannose for MPI-CDG) and organ transplantation (e.g., liver for MPI-CDG and heart for DOLK-CDG). However, for the majority of patients, only symptomatic and preventive treatments are in use. This constitutes a burden for patients, care-givers and ultimately the healthcare system. Innovative diagnostic approaches, in vitro and in vivo models and novel biomarkers have been developed that can lead to novel therapeutic avenues aiming to ameliorate the patients’ symptoms and lives. This review summarizes the advances in therapeutic approaches for CDG.


Subject(s)
Congenital Disorders of Glycosylation/diagnosis , Congenital Disorders of Glycosylation/genetics , Congenital Disorders of Glycosylation/therapy , Dietary Supplements , Genetic Therapy , Organ Transplantation , Animals , Biomarkers , Clinical Trials as Topic , Disease Models, Animal , Fucose/therapeutic use , Galactose/therapeutic use , Glycosylation , Humans , Mannose/therapeutic use
20.
Peptides ; 101: 17-24, 2018 03.
Article in English | MEDLINE | ID: mdl-29273518

ABSTRACT

D2A-Ala is a synthetic peptide that has been created by introducing mutations in the original D2A sequence, 130IQEGEEGRPKDDR142 of human urokinase receptor (uPAR). In vitro, D2A-Ala peptide displays strong anti-tumoural properties inhibiting EGF-induced chemotaxis, invasion and proliferation of a human fibrosarcoma cell line, HT 1080, and a human colorectal adenocarcinoma cell line, HT 29. D2A-Ala exerts its effects by preventing EGF receptor (EGFR) phosphorylation. To test D2A-Ala in vivo, this peptide was PEGylated generating polyethyleneglycol (PEG)-D2A-Ala peptide. PEGylation did not alter the inhibitory properties of D2A-Ala. Human tumour xenografts in the immunodeficient nude mice using HT 1080 and HT 29 cell lines showed that PEG-D2A-Ala significantly prevents tumour growth decreasing size, weight and density of tumours. The most efficient doses of the peptide were 5 and 10 mg/kg, thereby relevant for possible development of the peptide into a drug against cancer in particular tumours expressing EGFR.


Subject(s)
Adenosarcoma/drug therapy , Antineoplastic Agents/pharmacology , Chemotaxis/drug effects , Colorectal Neoplasms/drug therapy , Fibrosarcoma/drug therapy , Oligopeptides/pharmacology , Receptors, Urokinase Plasminogen Activator/chemistry , Adenosarcoma/metabolism , Adenosarcoma/pathology , Animals , Antineoplastic Agents/chemistry , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Female , Fibrosarcoma/metabolism , Fibrosarcoma/pathology , HT29 Cells , Humans , Mice , Mice, Nude , Neoplasm Invasiveness , Oligopeptides/chemistry , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...