Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167163, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38599261

ABSTRACT

PMM2-CDG (MIM # 212065), the most common congenital disorder of glycosylation, is caused by the deficiency of phosphomannomutase 2 (PMM2). It is a multisystemic disease of variable severity that particularly affects the nervous system; however, its molecular pathophysiology remains poorly understood. Currently, there is no effective treatment. We performed an RNA-seq based transcriptomic study using patient-derived fibroblasts to gain insight into the mechanisms underlying the clinical symptomatology and to identify druggable targets. Systems biology methods were used to identify cellular pathways potentially affected by PMM2 deficiency, including Senescence, Bone regulation, Cell adhesion and Extracellular Matrix (ECM) and Response to cytokines. Functional validation assays using patients' fibroblasts revealed defects related to cell proliferation, cell cycle, the composition of the ECM and cell migration, and showed a potential role of the inflammatory response in the pathophysiology of the disease. Furthermore, treatment with a previously described pharmacological chaperone reverted the differential expression of some of the dysregulated genes. The results presented from transcriptomic data might serve as a platform for identifying therapeutic targets for PMM2-CDG, as well as for monitoring the effectiveness of therapeutic strategies, including pharmacological candidates and mannose-1-P, drug repurposing.


Subject(s)
Congenital Disorders of Glycosylation , Fibroblasts , Phosphotransferases (Phosphomutases) , Humans , Congenital Disorders of Glycosylation/genetics , Congenital Disorders of Glycosylation/pathology , Congenital Disorders of Glycosylation/metabolism , Congenital Disorders of Glycosylation/drug therapy , Phosphotransferases (Phosphomutases)/genetics , Phosphotransferases (Phosphomutases)/metabolism , Phosphotransferases (Phosphomutases)/deficiency , Fibroblasts/metabolism , Fibroblasts/pathology , Transcriptome , Gene Expression Profiling , Cell Proliferation/genetics , Cell Proliferation/drug effects , Female , Male , Cell Movement/genetics , Cell Movement/drug effects
2.
Hum Mutat ; 43(10): 1430-1442, 2022 10.
Article in English | MEDLINE | ID: mdl-35789514

ABSTRACT

Different strategies are being investigated for treating PMM2-CDG, the most common congenital disorder of glycosylation. The use of pharmacochaperones (PCs) is one of the most promising. The present work characterizes the expression, stability, and enzymatic properties of 15 previously described clinical variants of the PMM2 protein, four novel variants, the Pmm2 mouse variant p.Phe115Leu, and its p.Phe119Leu human counterpart, with the aim of extending the potential use of pharmacochaperoning treatment. PMM2 variants were purified as stable homodimers, except for p.Asp65Gly, p.Ile120Thr, and p.Thr237Lys (no expression detected), p.Thr226Ser and p.Val231Met (aggregates), and p.Glu93Ala, p.Phe119Leu, and p.Phe115Leu (partial dissociated). Enzyme activity analyses identified severe variants and milder ones. Pure dimeric mutant proteins showed a reduction in thermal stability except for p.Asn216Asp. The thermal stability of all the unstable mutants was recovered in the presence of the PC compound VIII. This study adds to the list of destabilizing human variants amenable to rescue by small chemical compounds that increase the stability/activity of PMM2. The proposed platform can be reliably used for assessing the disease-causing effects of PMM2 missense variants, for assessing the correlation between genotype and phenotype, for confirming new clinical defects, and for identifying destabilizing mutations amenable to rescue by PCs.


Subject(s)
Congenital Disorders of Glycosylation , Phosphotransferases (Phosphomutases) , Animals , Congenital Disorders of Glycosylation/genetics , Glycosylation , Humans , Mice , Mutation , Phenotype , Phosphotransferases (Phosphomutases)/genetics
3.
Biochim Biophys Acta Gen Subj ; 1864(11): 129686, 2020 11.
Article in English | MEDLINE | ID: mdl-32712172

ABSTRACT

BACKGROUND: Mutations in the PMM2 gene cause phosphomannomutase 2 deficiency (PMM2; MIM# 212065), which manifests as a congenital disorder of glycosylation (PMM2-CDG). Mutant PMM2 leads to the reduced conversion of Man-6-P to Man-1-P, which results in low concentrations of guanosine 5'-diphospho-D-mannose, a nucleotide-activated sugar essential for the construction of protein oligosaccharide chains. To date the only therapeutic options are preventive and symptomatic. SCOPE OF REVIEW: This review covers the latest advances in the search for a treatment for PMM2-CDG. MAJOR CONCLUSIONS: Treatments based on increasing Man-1-P levels have been proposed, along with the administration of different mannose derivates, employing enzyme inhibitors or repurposed drugs to increase the synthesis of GDP-Man. A single repurposed drug that might alleviate a severe neurological symptom associated with the disorder is now in clinical use. Proof of concept also exists regarding the use of pharmacological chaperones and/or proteostatic regulators to increase the concentration of hypomorphic PMM2 mutant proteins. GENERAL SIGNIFICANCE: The ongoing challenges facing the discovery of drugs to treat this orphan disease are discussed.


Subject(s)
Congenital Disorders of Glycosylation/therapy , Phosphotransferases (Phosphomutases)/deficiency , Animals , Antisense Elements (Genetics)/therapeutic use , Congenital Disorders of Glycosylation/drug therapy , Congenital Disorders of Glycosylation/genetics , Congenital Disorders of Glycosylation/metabolism , Drug Discovery , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/therapeutic use , Glycosylation/drug effects , Humans , Mannose/analogs & derivatives , Mannose/therapeutic use , Phosphotransferases (Phosphomutases)/genetics , Phosphotransferases (Phosphomutases)/metabolism
4.
Hum Mutat ; 41(7): 1329-1338, 2020 07.
Article in English | MEDLINE | ID: mdl-32333439

ABSTRACT

Biallelic variants of the gene DNAJC12, which encodes a cochaperone, were recently described in patients with hyperphenylalaninemia (HPA). This paper reports the retrospective genetic analysis of a cohort of unsolved cases of HPA. Biallelic variants of DNAJC12 were identified in 20 patients (generally neurologically asymptomatic) previously diagnosed with phenylalanine hydroxylase (PAH) deficiency (phenylketonuria [PKU]). Further, mutations of DNAJC12 were identified in four carriers of a pathogenic variant of PAH. The genetic spectrum of DNAJC12 in the present patients included four new variants, two intronic changes c.298-2A>C and c.502+1G>C, presumably affecting the splicing process, and two exonic changes c.309G>T (p.Trp103Cys) and c.524G>A (p.Trp175Ter), classified as variants of unknown clinical significance (VUS). The variant p.Trp175Ter was detected in 83% of the mutant alleles, with 14 cases homozygous, and was present in 0.3% of a Spanish control population. Functional analysis indicated a significant reduction in PAH and its activity, reduced tyrosine hydroxylase stability, but no effect on tryptophan hydroxylase 2 stability, classifying the two VUS as pathogenic variants. Additionally, the effect of the overexpression of DNAJC12 on some destabilizing PAH mutations was examined and a mutation-specific effect on stabilization was detected suggesting that the proteostasis network could be a genetic modifier of PAH deficiency and a potential target for developing mutation-specific treatments for PKU.


Subject(s)
Phenylketonurias/genetics , Repressor Proteins/genetics , Adolescent , Adult , Alleles , Child , Child, Preschool , DNA Mutational Analysis , Exons , Humans , Infant , Infant, Newborn , Introns , RNA Splicing , Retrospective Studies , Spain
5.
PLoS One ; 14(2): e0212792, 2019.
Article in English | MEDLINE | ID: mdl-30802270

ABSTRACT

In Mexico, the increase in childhood obesity is alarming. Thus, improving the precision of its diagnosis is expected to impact on disease prevention. We estimated obesity prevalence by bioimpedance-based percent body fat (%BF) and body mass index (BMI) in 1061 girls and 1121 boys, from 3 to 17 years old. Multiple regressions and area under receiver operating curves (AUC) were used to determine the predictive value of BMI on %BF and percentile curves were constructed. Overall obesity prevalence estimated by %BF was 43.7%, and by BMI it was 20.1%; it means that the diagnosis by BMI underestimated around 50% of children diagnosed with obesity by %BF (≥30% for girls, ≥25% for boys). The fat mass excess is further underestimated in boys than in girls when using the standard BMI classification. The relationship between %BF and BMI was strong in school children and adolescents (all cases R2>0.70), but not in preschool children (girls R2 = 0.57, boys R2 = 0.23). AUCs showed greater discriminative power of BMI to detect %BF obesity in school children and adolescents (all cases AUC≥0.90) than in preschool children (girls AUC = 0.86; boys AUC = 0.70). Growth percentile charts showed that girls aged 9-17 years and boys aged 8-17 years presented fat excess from the 50th percentile and above. We suggested to change the BMI cut-off for them, considering values at the 75th percentile as overweight, and values at the 85th percentile as obesity, as previously recommended for Mexican children. Improving obesity diagnosis will allow greater efficiency when searching for comorbidities in clinical practice.


Subject(s)
Adipose Tissue , Body Mass Index , Pediatric Obesity , Adipose Tissue/pathology , Adipose Tissue/physiopathology , Adolescent , Child , Child, Preschool , Female , Humans , Male , Mexico/epidemiology , Pediatric Obesity/diagnosis , Pediatric Obesity/epidemiology , Pediatric Obesity/pathology , Pediatric Obesity/physiopathology , Prevalence , Sex Factors
6.
Mol Genet Metab ; 125(3): 266-275, 2018 11.
Article in English | MEDLINE | ID: mdl-30274917

ABSTRACT

Propionic acidemia (PA) is caused by mutations in the PCCA and PCCB genes, encoding α and ß subunits, respectively, of the mitochondrial enzyme propionyl-CoA carboxylase (PCC). Up to date, >200 pathogenic mutations have been identified, mostly missense defects. Genetic analysis in PA patients referred to the laboratory for the past 15 years identified 20 novel variants in the PCCA gene and 14 in the PCCB gene. 21 missense variants were predicted as probably disease-causing by different bioinformatics algorithms. Structural analysis in the available 3D model of the PCC enzyme indicated potential instability for most of them. Functional analysis in a eukaryotic system confirmed the pathogenic effect for the missense variants and for one amino acid deletion, as they all exhibited reduced or null PCC activity and protein levels compared to wild-type constructs. PCCB variants p.E168del, p.Q58P and p.I460T resulted in medium-high protein levels and no activity. Variants p.R230C and p.C712S in PCCA, and p.G188A, p.R272W and p.H534R in PCCB retained both partial PCC activity and medium-high protein levels. Available patients-derived fibroblasts carriers of some of these mutations were grown at 28 °C or 37 °C and a slight increase in PCC activity or protein could be detected in some cases at the folding-permissive conditions. Examination of available clinical data showed correlation of the results of the functional analysis with disease severity for most mutations, with some notable exceptions, confirming the notion that the final phenotypic outcome in PA is not easily predicted.


Subject(s)
Genetic Predisposition to Disease , Methylmalonyl-CoA Decarboxylase/genetics , Propionic Acidemia/genetics , Structure-Activity Relationship , Adolescent , Adult , Child , Child, Preschool , Female , Genetic Association Studies , Genotype , Humans , Infant , Infant, Newborn , Male , Methylmalonyl-CoA Decarboxylase/chemistry , Mitochondria/enzymology , Mitochondria/genetics , Mutation, Missense/genetics , Neonatal Screening , Propionic Acidemia/pathology , Protein Conformation , Protein Folding , Young Adult
7.
PLoS One ; 12(6): e0179456, 2017.
Article in English | MEDLINE | ID: mdl-28662078

ABSTRACT

Pathogenic mutations in DPAGT1 are manifested as two possible phenotypes: congenital disorder of glycosylation DPAGT1-CDG (also known as CDG-Ij), and limb-girdle congenital myasthenic syndrome (CMS) with tubular aggregates. UDP-N-acetylglucosamine-dolichyl-phosphate N-acetylglucosamine phosphotransferase (GPT), the protein encoded by DPAGT1, is an endoplasmic reticulum (ER)-resident protein involved in an initial step in the N-glycosylation pathway. The aim of the present study was to examine the effect of six variants in DPAGT1 detected in patients with DPAGT1-CDG, and the role of endoplasmic reticulum stress, as part of the search for therapeutic strategies to use against DPAGT1-CDG. The effect of the six mutations, i.e., c.358C>A (p.Leu120Met), c.791T>G (p.Val264Gly), c.901C>T (p.Arg301Cys), c.902G>A (p.Arg301His), c.1154T>G (p.Leu385Arg), and of the novel mutation c.329T>C (p.Phe110Ser), were examined via the analysis of DPAGT1 transcriptional profiles and GTP levels in patient-derived fibroblasts. In addition, the transient expression of different mutations was analysed in COS-7 cells. The results obtained, together with those of bioinformatic studies, revealed these mutations to affect the splicing process, the stability of GTP, or the ability of this protein to correctly localise in the ER membrane. The unfolded protein response (UPR; the response to ER stress) was found not to be active in patient-derived fibroblasts, unlike that seen in cells from patients with PMM2-CDG or DPM1-CDG. Even so, the fibroblasts of patients with DPAGT1-CDG seemed to be more sensitive to the stressor tunicamycin. The present work improves our knowledge of DPAGT1-CDG and provides bases for developing tailored splicing and folding therapies.


Subject(s)
Congenital Disorders of Glycosylation/genetics , Endoplasmic Reticulum Stress , Mutation , N-Acetylglucosaminyltransferases/physiology , Animals , COS Cells , Chlorocebus aethiops , Humans , Microscopy, Fluorescence , N-Acetylglucosaminyltransferases/genetics , Polymorphism, Single Nucleotide , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
8.
Hum Mutat ; 38(2): 160-168, 2017 02.
Article in English | MEDLINE | ID: mdl-27774737

ABSTRACT

The congenital disorder of glycosylation (CDG) due to phosphomannomutase 2 deficiency (PMM2-CDG), the most common N-glycosylation disorder, is a multisystem disease for which no effective treatment is available. The recent functional characterization of disease-causing mutations described in patients with PMM2-CDG led to the idea of a therapeutic strategy involving pharmacological chaperones (PC) to rescue PMM2 loss-of-function mutations. The present work describes the high-throughput screening, by differential scanning fluorimetry, of 10,000 low-molecular-weight compounds from a commercial library, to search for possible PCs for the enzyme PMM2. This exercise identified eight compounds that increased the thermal stability of PMM2. Of these, four compounds functioned as potential PCs that significantly increased the stability of several destabilizing and oligomerization mutants and also increased PMM activity in a disease model of cells overexpressing PMM2 mutations. Structural analysis revealed one of these compounds to provide an excellent starting point for chemical optimization since it passed tests based on a number of pharmacochemical quality filters. The present results provide the first proof-of-concept of a possible treatment for PMM2-CDG and describe a promising chemical structure as a starting point for the development of new therapeutic agents for this severe orphan disease.


Subject(s)
Congenital Disorders of Glycosylation/genetics , Phosphotransferases (Phosphomutases)/genetics , Alleles , Congenital Disorders of Glycosylation/drug therapy , Drug Discovery , Enzyme Activation , Fibroblasts/metabolism , Genotype , High-Throughput Screening Assays , Humans , Loss of Function Mutation , Molecular Targeted Therapy , Mutation , Phosphotransferases (Phosphomutases)/chemistry , Phosphotransferases (Phosphomutases)/isolation & purification , Protein Stability , Proteolysis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Small Molecule Libraries , Structure-Activity Relationship
9.
Hum Mutat ; 36(9): 851-60, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26014514

ABSTRACT

Congenital disorder of glycosylation type Ia (PMM2-CDG), the most common form of CDG, is caused by mutations in the PMM2 gene that reduce phosphomannomutase 2 (PMM2) activity. No curative treatment is available. The present work describes the functional analysis of nine human PMM2 mutant proteins frequently found in PMM2-CDG patients and also two murine Pmm2 mutations carried by the unique PMM2-CDG mouse model described to overcome embryonic lethality. The effects of the mutations on PMM2/Pmm2 stability, oligomerization, and enzyme activity were explored in an optimized bacterial system. The mutant proteins were associated with an enzymatic activity of up to 47.3% as compared with wild type (WT). Stability analysis performed using differential scanning fluorimetry and a bacterial transcription-translation-coupled system allowed the identification of several destabilizing mutations (p.V44A, p.D65Y, p.R123Q, p.R141H, p.R162W, p.F207S, p.T237M, p.C241S). Exclusion chromatography identified one mutation, p.P113L, that affected dimer interaction. Expression analysis of the p.V44A, p.D65Y, p.R162W, and p.T237M mutations in a eukaryotic expression system under permissive folding conditions showed the possibility of recovering their associated PMM2 activity. Together, the results suggest that some loss-of-function mutations detected in PMM2-CDG patients could be destabilizing, and therefore PMM2 activity could be, in certain cases, rescuable via the use of synergetic proteostasis modulators and/or chaperones.


Subject(s)
Congenital Disorders of Glycosylation/genetics , Congenital Disorders of Glycosylation/metabolism , Mutation , Phosphotransferases (Phosphomutases)/genetics , Phosphotransferases (Phosphomutases)/metabolism , Protein Folding , Animals , Enzyme Activation/genetics , Enzyme Stability/genetics , Fibroblasts , Gene Expression , Humans , Mice , Phosphotransferases (Phosphomutases)/chemistry , Protein Multimerization , Proteolysis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
10.
JIMD Rep ; 5: 59-70, 2012.
Article in English | MEDLINE | ID: mdl-23430918

ABSTRACT

Prospectively enrolled phenylketonuria patients (n=485) participated in an international Phase II clinical trial to identify the prevalence of a therapeutic response to daily doses of sapropterin dihydrochloride (sapropterin, KUVAN(®)). Responsive patients were then enrolled in two subsequent Phase III clinical trials to examine safety, ability to reduce blood Phenylalanine levels, dosage (5-20 mg/kg/day) and response, and bioavailability of sapropterin. We combined phenotypic findings in the Phase II and III clinical trials to classify study-related responsiveness associated with specific alleles and genotypes identified in the patients. We found that 17% of patients showed a response to sapropterin. The patients harbored 245 different genotypes derived from 122 different alleles, among which ten alleles were newly discovered. Only 16.3% of the genotypes clearly conferred a sapropterin-responsive phenotype. Among the different PAH alleles, only 5% conferred a responsive phenotype. The responsive alleles were largely but not solely missense mutations known to or likely to cause misfolding of the PAH subunit. However, the metabolic response was not robustly predictable from the PAH genotypes, based on the study design adopted for these clinical trials, and accordingly it seems prudent to test each person for this phenotype with a standardized protocol.

11.
Mol Genet Metab ; 104(3): 249-54, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21803624

ABSTRACT

Phenylketonuria (PKU), a Mendelian autosomal recessive phenotype (OMIM 261600), is an inborn error of metabolism causing impaired postnatal cognitive development in the absence of treatment. We used the Pah(enu2/enu2) PKU mouse model to study oral enzyme substitution therapy with various chemically modified formulations of phenylalanine ammonia lyase (Av-p.C503S/p.C565S/p.F18A PAL). In vivo studies with the most therapeutically effective formulation (5kDa PEG-Av-p.C503S/p.C565S/p.F18A PAL) revealed that this conjugate, given orally, yielded statistically significant (p=0.0029) and therapeutically relevant reduction (~40%) in plasma phenylalanine (Phe) levels. Phe reduction occurred in a dose- and loading-dependent manner; sustained clinically and statistically significant reduction of plasma Phe levels was observed with treatment ranging between 0.3 IU and 9 IU and with more frequent and smaller dosings. Oral PAL therapy could potentially serve as an adjunct therapy, perhaps with dietary treatment, and will work independently of phenylalanine hydroxylase (PAH), correcting such forms of hyperphenylalaninemias regardless of the PAH mutations carried by the patient.


Subject(s)
Phenylalanine Ammonia-Lyase/therapeutic use , Phenylalanine/blood , Phenylketonurias/drug therapy , Administration, Oral , Alginates , Anabaena variabilis/enzymology , Analysis of Variance , Animals , Basidiomycota/enzymology , Chitosan , Dextran Sulfate , Dose-Response Relationship, Drug , Glucuronic Acid , Hexuronic Acids , Mice , Nanoparticles , Phenylalanine Ammonia-Lyase/administration & dosage , Phenylalanine Ammonia-Lyase/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism
12.
J Inherit Metab Dis ; 34(4): 929-39, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21541725

ABSTRACT

Deficiency of phosphomannomutase (PMM2, MIM#601785) is the most common congenital disorder of glycosylation. Herein we report the genetic analysis of 22 Spanish PMM2 deficient patients and the functional analysis of 14 nucleotide changes in a prokaryotic expression system in order to elucidate their molecular pathogenesis. PMM2 activity assay revealed the presence of six protein changes with no enzymatic activities (p.R123Q, p.R141H, p.F157S, p.P184T, p.F207S and p.D209G) and seven mild protein changes with residual activities ranging from 16 to 54% (p.L32R, p.V44A p.D65Y, p.P113L p.T118S, p.T237M and p.C241S) and also one variant change with normal activity (p.E197A). The results obtained from Western blot analysis, degradation time courses of 11 protein changes and structural analysis of the PMM2 protein, suggest that the loss-of-function of most mutant proteins is based on their increased susceptibility to degradation or aggregation compared to the wild type protein, considering PMM2 deficiency as a conformational disease. We have identified exclusively catalytic protein change (p.D209G), catalytic protein changes affecting protein stability (p.R123Q and p.R141H), two protein changes disrupting the dimer interface (p.P113L and p.T118S) and several misfolding changes (p.L32R, p.V44A, p.D65Y, p.F157S, p.P184T, p.F207S, p.T237M and p.C241S). Our current work opens a promising therapeutic option using pharmacological chaperones to revert the effect of the characterized misfolding mutations identified in a wide range of PMM2 deficient patients.


Subject(s)
Congenital Disorders of Glycosylation/genetics , Mutation, Missense/physiology , Phosphotransferases (Phosphomutases)/genetics , Cell Line , Congenital Disorders of Glycosylation/metabolism , Congenital Disorders of Glycosylation/pathology , DNA Mutational Analysis , Enzyme Stability/genetics , Gene Expression Profiling , Gene Expression Regulation, Enzymologic/genetics , Humans , Phosphotransferases (Phosphomutases)/deficiency , Phosphotransferases (Phosphomutases)/metabolism , Protein Folding
13.
Biochim Biophys Acta ; 1802(11): 959-67, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20696242

ABSTRACT

An increased reactive oxygen species (ROS) production and apoptosis rate have been associated with several disorders involved in cobalamin metabolism, including isolated methylmalonic aciduria (MMA) cblB type and MMA combined with homocystinuria (MMAHC) cblC type. Given the relevance of p38 and JNK kinases in stress-response, their activation in fibroblasts from a spectrum of patients (mut, cblA, cblB, cblC and cblE) was analyzed revealing an increased expression of the phosphorylated-forms, specially in cblB and cblC cell lines that presented the highest ROS and apoptosis levels. To gain further insight into the molecular mechanisms responsible for the enhanced apoptotic process observed in cblB and cblC fibroblasts, we evaluated the expression pattern of 84 apoptosis-related genes by quantitative real-time PCR. An elevated number of pro-apoptotic genes were overexpressed in cblC cells showing a higher rate of apoptosis compared to cblB and control samples. Additionally, apoptosis appears to be mainly triggered through the extrinsic pathway in cblC, while the intrinsic pathway was primarily activated in cblB cells. The differences observed regarding the apoptosis rate and preferred pathway between cblB and cblC patients, who both built up methylmalonic acid, might be explained by the accumulated homocysteine in the cblC group. The loss of MMACHC function in cblC patients might be partially responsible for the oxidative stress and apoptosis processes observed in these cell lines. Our results suggest that ROS production may represent a genetic modifier of the phenotype and support the potential of using antioxidants as a novel therapeutic strategy to improve the severe neurological outcome of these rare diseases.


Subject(s)
Amino Acid Metabolism, Inborn Errors/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis/genetics , Gene Expression Profiling , Homocystinuria/genetics , Amino Acid Metabolism, Inborn Errors/classification , Amino Acid Metabolism, Inborn Errors/metabolism , Apoptosis Regulatory Proteins/metabolism , Blotting, Western , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Fibroblasts/metabolism , Homocystinuria/classification , Homocystinuria/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Methylmalonic Acid/metabolism , Mutation , Oxidoreductases , Phosphoproteins/metabolism , Phosphorylation , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
14.
Hum Mutat ; 31(9): 1033-42, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20556797

ABSTRACT

ATP:cob(I)alamin adenosyltransferase (ATR, E.C.2.5.1.17) converts reduced cob(I)alamin to the adenosylcobalamin cofactor. Mutations in the MMAB gene encoding ATR are responsible for the cblB type methylmalonic aciduria. Here we report the functional analysis of five cblB mutations to determine the underlying molecular basis of the dysfunction. The transcriptional profile along with minigenes analysis revealed that c.584G>A, c.349-1G>C, and c.290G>A affect the splicing process. Wild-type ATR and the p.I96T (c.287T>C) and p.R191W (c.571C>T) mutant proteins were expressed in a prokaryote and a eukaryotic expression systems. The p.I96T protein was enzymatically active with a K(M) for ATP and K(D) for cob(I)alamin similar to wild-type enzyme, but exhibited a 40% reduction in specific activity. Both p.I96T and p.R191W mutant proteins are less stable than the wild-type protein, with increased stability when expressed under permissive folding conditions. Analysis of the oligomeric state of both mutants showed a structural defect for p.I96T and also a significant impact on the amount of recovered mutant protein that was more pronounced for p.R191W that, along with the structural analysis, suggest they might be misfolded. These results could serve as a basis for the implementation of pharmacological therapies aimed at increasing the residual activity of this type of mutations.


Subject(s)
Mutation/genetics , Alkyl and Aryl Transferases/genetics , Amino Acid Metabolism, Inborn Errors/enzymology , Amino Acid Metabolism, Inborn Errors/genetics , Cell Line , Child, Preschool , DNA Mutational Analysis , Female , Genome, Human/genetics , Humans , Infant , Infant, Newborn , Kinetics , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutation, Missense/genetics , Protein Structure, Quaternary , Protein Structure, Secondary , RNA Splicing/genetics , Time Factors
15.
Mol Genet Metab ; 99(1): 4-9, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19793667

ABSTRACT

Phenylalanine ammonia lyase (PAL) has long been recognized as a potential enzyme replacement therapeutic for treatment of phenylketonuria. However, various strategies for the oral delivery of PAL have been complicated by the low intestinal pH, aggressive proteolytic digestion and circulation time in the GI tract. In this work, we report 3 strategies to address these challenges. First, we used site-directed mutagenesis of a chymotrypsin cleavage site to modestly improve protease resistance; second, we used silica sol-gel material as a matrix to demonstrate that a silica matrix can provide protection to entrapped PAL proteins against intestinal proteases, as well as a low pH of 3.5; finally, we demonstrated that PEGylation of AvPAL surface lysines can reduce the inactivation of the enzyme by trypsin.


Subject(s)
Bacterial Proteins/therapeutic use , Enzyme Replacement Therapy/methods , Phenylalanine Ammonia-Lyase/therapeutic use , Phenylketonurias/drug therapy , Administration, Oral , Anabaena variabilis/enzymology , Bacterial Proteins/administration & dosage , Bacterial Proteins/metabolism , Binding Sites/genetics , Chymotrypsin/metabolism , Enzyme Stability , Hot Temperature , Humans , Hydrogen-Ion Concentration , Injections , Models, Molecular , Mutagenesis, Site-Directed , Phenylalanine Ammonia-Lyase/genetics , Phenylalanine Ammonia-Lyase/metabolism , Polyethylene Glycols/chemistry , Protein Engineering/methods , Protein Multimerization , Protein Structure, Quaternary , Silicon Dioxide/chemistry , Structure-Activity Relationship , Technology, Pharmaceutical/methods , Trypsin/metabolism
16.
Proc Natl Acad Sci U S A ; 105(52): 20894-9, 2008 Dec 30.
Article in English | MEDLINE | ID: mdl-19095795

ABSTRACT

Phenylketonuria (PKU) is a metabolic disorder, in which loss of phenylalanine hydroxylase activity results in neurotoxic levels of phenylalanine. We used the Pah(enu2/enu2) PKU mouse model in short- and long-term studies of enzyme substitution therapy with PEGylated phenylalanine ammonia lyase (PEG-PAL conjugates) from 4 different species. The most therapeutically effective PAL (Av, Anabaena variabilis) species was one without the highest specific activity, but with the highest stability; indicating the importance of protein stability in the development of effective protein therapeutics. A PEG-Av-p.C503S/p.C565S-PAL effectively lowered phenylalanine levels in both vascular space and brain tissue over a >90 day trial period, resulting in reduced manifestations associated with PKU, including reversal of PKU-associated hypopigmentation and enhanced animal health. Phenylalanine reduction occurred in a dose- and loading-dependent manner, and PEGylation reduced the neutralizing immune response to the enzyme. Human clinical trials with PEG-Av-p.C503S/p.C565S-PAL as a treatment for PKU are underway.


Subject(s)
Anabaena variabilis/enzymology , Antineoplastic Agents/pharmacology , Bacterial Proteins/pharmacology , Phenylalanine Ammonia-Lyase/pharmacology , Phenylketonurias/drug therapy , Polyethylene Glycols , Recombinant Proteins/pharmacology , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Bacterial Proteins/adverse effects , Bacterial Proteins/therapeutic use , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Enzyme Stability/physiology , Humans , Mice , Mice, Mutant Strains , Organ Specificity/drug effects , Phenylalanine/metabolism , Phenylalanine Ammonia-Lyase/adverse effects , Phenylalanine Ammonia-Lyase/therapeutic use , Phenylketonurias/metabolism , Recombinant Proteins/adverse effects , Recombinant Proteins/therapeutic use
17.
J Mol Biol ; 380(4): 623-35, 2008 Jul 18.
Article in English | MEDLINE | ID: mdl-18556022

ABSTRACT

We have recently observed promising success in a mouse model for treating the metabolic disorder phenylketonuria with phenylalanine ammonia lyase (PAL) from Rhodosporidium toruloides and Anabaena variabilis. Both molecules, however, required further optimization in order to overcome problems with protease susceptibility, thermal stability, and aggregation. Previously, we optimized PAL from R. toruloides, and in this case we reduced aggregation of the A. variabilis PAL by mutating two surface cysteine residues (C503 and C565) to serines. Additionally, we report the structural and biochemical characterization of the A. variabilis PAL C503S/C565S double mutant and carefully compare this molecule with the R. toruloides engineered PAL molecule. Unlike previously published PAL structures, significant electron density is observed for the two active-site loops in the A. variabilis C503S/C565S double mutant, yielding a complete view of the active site. Docking studies and N-hydroxysuccinimide-biotin binding studies support a proposed mechanism in which the amino group of the phenylalanine substrate is attacked directly by the 4-methylidene-imidazole-5-one prosthetic group. We propose a helix-to-loop conformational switch in the helices flanking the inner active-site loop that regulates accessibility of the active site. Differences in loop stability among PAL homologs may explain the observed variation in enzyme efficiency, despite the highly conserved structure of the active site. A. variabilis C503S/C565S PAL is shown to be both more thermally stable and more resistant to proteolytic cleavage than R. toruloides PAL. Additional increases in thermal stability and protease resistance upon ligand binding may be due to enhanced interactions among the residues of the active site, possibly locking the active-site structure in place and stabilizing the tetramer. Examination of the A. variabilis C503S/C565S PAL structure, combined with analysis of its physical properties, provides a structural basis for further engineering of residues that could result in a better therapeutic molecule.


Subject(s)
Anabaena variabilis/enzymology , Bacterial Proteins/chemistry , Phenylalanine Ammonia-Lyase/chemistry , Protein Structure, Tertiary , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Crystallography, X-Ray , Enzyme Stability , Gene Duplication , Hydrogen-Ion Concentration , Ligands , Mice , Models, Molecular , Molecular Sequence Data , Peptide Hydrolases/metabolism , Phenylalanine Ammonia-Lyase/genetics , Phenylalanine Ammonia-Lyase/metabolism , Point Mutation , Protein Engineering , Protein Structure, Quaternary , Temperature
18.
Cochabamba; s.n; 200803. 55 p. ^empastado.
Thesis in Spanish | LIBOCS, LILACS, LIBOSP | ID: biblio-1308245

ABSTRACT

La puericultura en pediatría es una rama importante d la que depende no solo la salud preventiva del niño, sino también es la base para lograr la confianza de los padres en pediatría de su niño, al escuchar y orientarles sobre aspectos que van más allá de una plática meramente terapéutica, el profesional debe suministrar educación e información, acerca de aspectos tan básicos como valiosos, como ser un adecuado desarrollo y crecimiento, una alimentación apropiada, así como también la prevención de enfermedades u accidentes.En ocasiones la consulta numerosa, no permite al pediatra disponer de tiempo para dedicar a un aspecto tan delicado, como es la enseñanza de la crianza de un niño.En los grandes hospitales de especialidad donde nos formamos aprendemos a tratar patologías y saber reconocerlas, pero es también necesario aprender a tratar al niño en su contexto sano , saber brindar a los padres un consejo veraz, científico, satisfacer sus inquietudes y sus dudas. Se decide realizar este estudio con la finalidad de saber cuál s el conocimiento del pediatra en formación, en éste campo, así como también si estos conocimientos son aplicados en la práctica de su formación.


Subject(s)
Child Care , Pediatrics
19.
Mol Genet Metab ; 91(4): 325-34, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17560821

ABSTRACT

Protein and peptide therapeutics are of growing importance as medical treatments but can frequently induce an immune response. This work describes the combination of complementary approaches to map the potential immunogenic regions of the yeast Rhodosporidium toruloides phenylalanine ammonia-lyase (PAL, EC 4.3.1.5) and to engineer the protein as a human therapeutic agent for the treatment of phenylketonuria (PKU), an inherited metabolic disorder. The identification of B and T cell epitopes on the PAL protein was performed by computational predictions based on the antigenicity and hydrophilicity of proteins, as well as by experimental epitope mapping using a PepSpots peptide array (Jerini AG). Human T cell epitope mapping was performed by applying the computational EpiMatrix algorithm (EpiVax, Inc.) for MHC Class I and Class II associated T cell epitopes on PAL, which predicts which sequences are associated with binding to several different HLA alleles, a requirement for antigen presentation and subsequent primary immune response. By chemical modification through PEGylation of surface lysine residues, it is possible to cover the immunogenic regions of a protein. To evaluate this strategy, we used mass spectrometry to determine which of the immunogenic epitopes are covered by the covalent PEGylation modification strategy. This approach has allowed us to determine whether additional lysines are needed in specific residue locations, or whether certain lysine residues can be removed in order to accomplish complete molecular coverage of the therapeutic enzyme.


Subject(s)
Phenylalanine Ammonia-Lyase/metabolism , Phenylalanine Ammonia-Lyase/therapeutic use , Phenylketonurias/drug therapy , B-Lymphocytes/immunology , Epitopes/analysis , Fungal Proteins/metabolism , Fungal Proteins/therapeutic use , Humans , Models, Molecular , Peptide Fragments/immunology , Phenylalanine Ammonia-Lyase/chemistry , Phenylketonurias/enzymology , Polyethylene Glycols , Protein Conformation , Rhodotorula/enzymology , T-Lymphocytes/immunology
20.
Mol Genet Metab ; 86 Suppl 1: S22-6, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16165390

ABSTRACT

Phenylketonuria (PKU) is an autosomal recessive genetic disorder in which mutations in the phenylalanine-4-hydroxylase (PAH) gene result in an inactive enzyme (PAH, EC 1.14.16.1). The effect is an inability to metabolize phenylalanine (Phe), translating into elevated levels of Phe in the bloodstream (hyperphenylalaninemia). If therapy is not implemented at birth, mental retardation can occur. PKU patients respond to treatment with a low-phenylalanine diet, but compliance with the diet is difficult, therefore the development of alternative treatments is desirable. Enzyme substitution therapy with a recombinant phenylalanine ammonia lyase (PAL) is currently being explored. This enzyme converts Phe to the harmless metabolites, trans-cinnamic acid and trace ammonia. Taken orally and when non-absorbable and protected, PAL lowers plasma Phe in mutant hyperphenylalaninemic mouse models. Subcutaneous administration of PAL results in more substantial lowering of plasma and significant reduction in brain Phe levels, however the metabolic effect is not sustained following repeated injections due to an immune response. We have chemically modified PAL by pegylation to produce a protected form of PAL that possesses better specific activity, prolonged half-life, and reduced immunogenicity in vivo. Subcutaneous administration of pegylated molecules to PKU mice has the desired metabolic response (prolonged reduction in blood Phe levels) with greatly attenuated immunogenicity.


Subject(s)
Phenylalanine Ammonia-Lyase/therapeutic use , Phenylketonurias/drug therapy , Animals , Humans , Phenylalanine/metabolism , Phenylalanine Hydroxylase/genetics , Phenylketonurias/genetics , Recombinant Proteins/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...