Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
JIMD Rep ; 27: 17-26, 2016.
Article in English | MEDLINE | ID: mdl-26404456

ABSTRACT

Cellular phenotyping of human dermal fibroblasts (HDFs) from patients with inherited diseases provides invaluable information for diagnosis, disease aetiology, prognosis and assessing of treatment options. Here we present a cell phenotyping protocol using image cytometry that combines measurements of crucial cellular and mitochondrial parameters: (1) cell number and viability, (2) thiol redox status (TRS), (3) mitochondrial membrane potential (MMP) and (4) mitochondrial superoxide levels (MSLs). With our protocol, cell viability, TRS and MMP can be measured in one small cell sample and MSL on a parallel one. We analysed HDFs from healthy individuals after treatment with various concentrations of hydrogen peroxide (H2O2) for different intervals, to mimic the physiological effects of oxidative stress. Our results show that cell number, viability, TRS and MMP decreased, while MSL increased both in a time- and concentration-dependent manner. To assess the use of our protocol for analysis of HDFs from patients with inherited diseases, we analysed HDFs from two patients with very long-chain acyl-CoA dehydrogenase (VLCAD) deficiency (VLCADD), one with a severe clinical phenotype and one with a mild one. HDFs from both patients displayed increased MSL without H2O2 treatment. Treatment with H2O2 revealed significant differences in MMP and MSL between HDFs from the mild and the severe patient. Our results establish the capacity of our protocol for fast analysis of cellular and mitochondrial parameters by image cytometry in HDFs from patients with inherited metabolic diseases.

2.
Free Radic Res ; 48(2): 168-79, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24151936

ABSTRACT

Even though the deleterious effects of increased reactive oxygen species (ROS) levels have been implicated in a variety of neurodegenerative disorders, the triggering events that lead to the increased ROS and successive damages are still ill-defined. Mitochondria are the key organelles controlling the ROS balance, being their main source and also counteracting them by the action of the ROS scavenging system. Mitochondria, moreover, control the presence of ROS-damaged proteins by action of the protein quality control (PQC) system. One of its components is the mitochondrial chaperone Hsp60 assisting the folding of a subset of mitochondrial matrix proteins. Mutations in Hsp60 cause a late onset form of the neurodegenerative disease hereditary spastic paraplegia (SPG13). In this study, we aimed to address the molecular consequences of Hsp60 shortage. We here demonstrate that a heterozygous knockout Hsp60 model that recapitulates features of the human disease and exhibits increased oxidative stress in neuronal tissues. Moreover, we indicate that the increase of ROS is, at least in part, due to impaired folding of the manganese superoxide dismutase (MnSOD), a key antioxidant enzyme. We observed that the Hsp60 and MnSOD proteins interact. Based on these results, we propose that MnSOD is a substrate of the Hsp60 folding machinery and that under conditions of diminished availability of Hsp60, MnSOD is impaired in reaching the native state. This suggests a possible link between Hsp60-dependent PQC and the ROS scavenging systems that may have the function to increase ROS production under conditions of folding stress.


Subject(s)
Chaperonin 60/physiology , Mitochondrial Proteins/physiology , Protein Folding , Superoxide Dismutase/metabolism , Animals , Cerebral Cortex/enzymology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/enzymology , Oxidative Stress , Reactive Oxygen Species/metabolism , Superoxide Dismutase/genetics , Transcription, Genetic
3.
Clin Genet ; 83(1): 44-52, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22168581

ABSTRACT

Familial neurohypophyseal diabetes insipidus (FNDI) typically presents with age-dependent penetrance and autosomal dominant inheritance caused by missense variations in one allele of the AVP gene encoding the arginine vasopressin (AVP) prohormone. We present the molecular genetic characteristics underlying an unusual form of FNDI occurring with very early onset and seemingly autosomal recessive inheritance. By DNA amplification and sequencing, we identified a novel variant allele of the AVP gene carrying a 10,396 base pair deletion involving the majority of the AVP gene as well as its regulatory sequences in the intergenic region between the AVP and the OXT gene, encoding the oxytocin prohormone. We found two chromosomes carrying the deletion in affected family members and one in unaffected family members suspected to transmit the deleted allele. Whole-genome array analysis confirmed the results and excluded the presence of any additional major pathogenic abnormalities. The deletion is predicted to abolish the transcription of the AVP gene, thus the fact that family members heterozygous for the deletion remain healthy argues, in general, against haploinsufficiency as the pathogenic mechanism FNDI. Accordingly, our data is strong support to the prevailing idea that dominant inheritance of FNDI is due to a dominant-negative effect exerted by variant AVP prohormone.


Subject(s)
Diabetes Insipidus, Neurogenic/genetics , Neurophysins/genetics , Protein Precursors/genetics , Sequence Deletion/genetics , Vasopressins/genetics , Age of Onset , Alleles , Diabetes Insipidus, Neurogenic/physiopathology , Female , Genes, Recessive , Heterozygote , Humans , Infant , Infant, Newborn , Male , Mutation , Pedigree , Pregnancy
4.
Clin Pharmacol Ther ; 92(5): 642-50, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22990751

ABSTRACT

Previous studies have demonstrated that the pharmacokinetic profile of erythromycin, a probe for CYP3A4 activity, is affected by inhibitors or inducers of hepatic solute carriers. We hypothesized that these interactions are mediated by OATP1B1 (gene symbol, SLCO1B1), a polypeptide expressed on the basolateral surface of hepatocytes. Using stably transfected Flp-In T-Rex293 cells, erythromycin was found to be a substrate for OATP1B1*1A (wild type) with a Michaelis-Menten constant of ~13 µmol/l, and that its transport was reduced by ~50% in cells expressing OATP1B1*5 (V174A). Deficiency of the ortholog transporter Oatp1b2 in mice was associated with a 52% decrease in the metabolic rate of erythromycin (P = 0.000043). In line with these observations, in humans the c.521T>C variant in SLCO1B1 (rs4149056), encoding OATP1B1*5, was associated with a decline in erythromycin metabolism (P = 0.0072). These results suggest that impairment of OATP1B1 function can alter erythromycin metabolism, independent of changes in CYP3A4 activity.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Erythromycin/pharmacokinetics , Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transporters/genetics , Adult , Aged , Aged, 80 and over , Animals , Biological Transport , Cell Line , Cytochrome P-450 CYP3A/metabolism , Female , Humans , Liver-Specific Organic Anion Transporter 1 , Male , Mice , Mice, Knockout , Middle Aged , Organic Anion Transporters/metabolism , Polymorphism, Genetic
5.
Mol Genet Metab ; 100(2): 155-62, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20371198

ABSTRACT

Short-chain acyl-CoA dehydrogenase deficiency (SCADD) is a rare inherited disorder of the mitochondrial beta-oxidation of fatty acids. Patients with SCADD present mainly with symptoms of neuromuscular character. In order to investigate factors involved in the pathogenesis, we studied a disease-associated variant of the SCAD protein (p.Arg83Cys, c.319C>T), which is known to compromise SCAD protein folding. We investigated the consequences of overexpressing the misfolded mitochondrial protein, and thus determined whether the misfolded p.Arg83Cys SCAD proteins can elicit a toxic reaction. Human astrocytes were transiently transfected with either wild-type or p.Arg83Cys encoding cDNA, and analyzed for insoluble proteins/aggregate-formation, alterations in mitochondrial morphology, and for the presence of reactive oxygen species (ROS) in the mitochondria. The majority of cells overexpressing the p.Arg83Cys SCAD variant protein presented with an altered mitochondrial morphology of a grain-like structure, whereas the majority of the cells overexpressing wild-type SCAD presented with a normal thread-like mitochondrial reticulum. We found this grain-like structure to be associated with an increased amount of ROS. The mitochondrial morphology change was partly alleviated by addition of the mitochondrial targeted antioxidant MitoQ, indicating a ROS-induced mitochondrial fission. We therefore propose that SCAD misfolding leads to production of ROS, which in turn leads to fission and a grain-like structure of the mitochondrial reticulum. This finding indicates a toxic response elicited by misfolded p.Arg83Cys SCAD proteins.


Subject(s)
Butyryl-CoA Dehydrogenase/chemistry , Lipid Metabolism, Inborn Errors/genetics , Mitochondria/genetics , Oxidative Stress/genetics , Protein Folding , Proteostasis Deficiencies/genetics , Astrocytes/enzymology , Butyryl-CoA Dehydrogenase/deficiency , Butyryl-CoA Dehydrogenase/genetics , Cell Line , Humans , Mitochondria/metabolism , Mitochondria/ultrastructure , Reactive Oxygen Species/metabolism , Transfection
6.
Neuroscience ; 153(2): 474-82, 2008 May 02.
Article in English | MEDLINE | ID: mdl-18378094

ABSTRACT

The mitochondrial chaperonin heat shock protein 60 (Hsp60) assists the folding of a subset of proteins localized in mitochondria and is an essential component of the mitochondrial protein quality control system. Mutations in the HSPD1 gene that encodes Hsp60 have been identified in patients with an autosomal dominant form of hereditary spastic paraplegia (SPG13), a late-onset neurodegenerative disorder characterized by a progressive paraparesis of the lower limbs. The disease-associated Hsp60-(p.Val98Ile) protein, encoded by the c.292G>A HSPD1 allele, has reduced chaperonin activity, but how its expression affects mitochondrial functions has not been investigated. We have studied mitochondrial function and expression of genes encoding mitochondrial chaperones and proteases in a human lymphoblastoid cell line and fibroblast cells from a patient who is heterozygous for the c.292G>A HSPD1 allele. We found that both the c.292G>A RNA transcript and the corresponding Hsp60-(p.Val98Ile) protein were present at comparable levels to their wild-type counterparts in SPG13 patient cells. Compared with control cells, we found no significant cellular or mitochondrial dysfunctions in SPG13 patient cells by assessing the mitochondrial membrane potential, cell viability, and sensitivity toward oxidative stress. However, a decreased expression of the mitochondrial protein quality control proteases Lon and ClpP, both at the RNA and protein level, was demonstrated in SPG13 patient cells. We propose that decreased levels of mitochondrial proteases Lon and ClpP may allow Hsp60 substrate proteins to go through more folding attempts instead of being prematurely degraded, thereby supporting productive folding in cells with reduced Hsp60 chaperonin activity. In conclusion, our studies with SPG13 patient cells expressing the functionally impaired mutant Hsp60 chaperonin suggest that reduction of the degradative activity of the protein quality control system may represent a previously unrecognized cellular adaptation to reduced chaperone function.


Subject(s)
Endopeptidase Clp/genetics , Mitochondria/enzymology , Protease La/genetics , Spastic Paraplegia, Hereditary/genetics , Alleles , Blotting, Western , Cell Line , Cell Survival , Cells, Cultured , Endopeptidase Clp/metabolism , Fibroblasts , HSP70 Heat-Shock Proteins/metabolism , Humans , Immunohistochemistry , Membrane Potentials , Molecular Chaperones/metabolism , Protease La/metabolism , RNA/biosynthesis , RNA/isolation & purification , Spastic Paraplegia, Hereditary/metabolism
7.
Mol Psychiatry ; 11(12): 1126-38, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16924267

ABSTRACT

Linkage studies suggest that chromosome 22q12-13 may contain one or more shared susceptibility genes for schizophrenia (SZ) and bipolar affective disorder (BPD). In a Faeroese sample, we previously reported association between microsatellite markers located at 22q13.31-qtel and both disorders. The present study reports an association analysis across five genes (including 14 single nucleotide and two microsatellite polymorphisms) in this interval using a case-control sample of 162 BPD, 103 SZ patients and 200 controls. The bromodomain-containing 1 gene (BRD1), which encodes a putative regulator of transcription showed association with both disorders with minimal P-values of 0.0046 and 0.00001 for single marker and overall haplotype analysis, respectively. A specific BRD1 2-marker 'risk' haplotype showed a frequency of approximately 10% in the combined case group versus approximately 1% in controls (P-value 2.8 x 10(-7)). Expression analysis of BRD1 mRNA revealed widespread expression in mammalian brain tissue, which was substantiated by immunohistochemical detection of BRD1 protein in the nucleus, perikaryal cytosol and proximal dendrites of the neurons in the adult rat, rabbit and human CNS. Quantitative mRNA analysis in developing fetal pig brain revealed spatiotemporal differences with high expression at early embryonic stages, with intense nuclear and cytosolar immunohistochemical staining of the neuroepithelial layer and early neuroblasts, whilst more mature neurons at later embryonic stages had less nuclear staining. The results implicate BRD1 with SZ and BPD susceptibility and provide evidence that suggests a role for BRD1 in neurodevelopment.


Subject(s)
Bipolar Disorder/genetics , Chromosomes, Human, Pair 22 , Genetic Linkage , Genetic Predisposition to Disease , Nuclear Proteins/genetics , Schizophrenia/genetics , Animals , Bipolar Disorder/metabolism , Bipolar Disorder/pathology , Brain/embryology , Brain/pathology , Case-Control Studies , Female , Gene Expression Profiling , Gene Expression Regulation , Genotype , Histone Acetyltransferases , Histone Chaperones , Humans , Male , Microsatellite Repeats , Nuclear Proteins/biosynthesis , Polymorphism, Single Nucleotide , Rabbits , Rats , Schizophrenia/metabolism , Schizophrenia/pathology , Swine
8.
Theriogenology ; 64(7): 1536-45, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-15935461

ABSTRACT

The purpose of our work was to establish an efficient protocol for activation of porcine cytoplast-fibroblast constructs produced by the handmade cloning technique. Firstly, we investigated a combined electrical and chemical activation protocol for parthenogenetic development of in vitro matured zona-free oocytes. Oocytes were activated by one 80 micros pulse and subsequently cultured in cytochalasin B and cycloheximide. Developmental rates of blastocysts from activated oocytes were 49+/-1 and 40+/-2%, when using one 80 micros pulse of 0.85 or 1.25 kV/cm, respectively. The activation procedure was further confirmed by a simultaneous re-fusion and activation of bisected oocytes, resulting in a blastocyst rate of 41+/-8%. Secondly, the activation protocol was applied in the handmade cloning technique. In vitro matured zona-free porcine oocytes were bisected and halves containing no chromatin, i.e. the cytoplasts, were selected. Reconstructed embryos were produced by a two-step fusion procedure. At the first step, one cytoplast was fused to one fibroblast by one 80 micros pulse of 1.25 kV/cm. After 1h, the cytoplast-fibroblast pair and another cytoplast were fused and activated simultaneously by one 80 micros pulse of 0.85 kV/cm, and subsequently cultured in cytochalasin B and cycloheximide. The development of reconstructed embryos to the blastocyst stage was in average 21+/-4%, and total blastocyst cell counts were in average 48+/-3. Thus, the combined electrical and chemical activation procedure resulted in efficient blastocyst development in the handmade cloning technique.


Subject(s)
Blastocyst , Cloning, Organism/veterinary , Oocytes/physiology , Swine/embryology , Animals , Cloning, Organism/methods , Cycloheximide/administration & dosage , Cytochalasin B/administration & dosage , Electric Stimulation , Female
9.
Reprod Fertil Dev ; 16(3): 315-8, 2004.
Article in English | MEDLINE | ID: mdl-15304204

ABSTRACT

Recently, a zona-free technique for bovine somatic cell nuclear transfer (NT) with no requirement for micromanipulation (i.e. hand-made cloning (HMC)) has been described. The present study demonstrates the application of the HMC technique in the production of transgenic porcine blastocysts. In vitro-matured zona-free porcine oocytes were bisected manually using a microblade and halves containing no chromatin (i.e. the cytoplasts) were selected. Two cytoplasts were electrofused with one transgenic fibroblast expressing enhanced green fluorescent protein and reconstructed embryos were activated in calcium ionophore (A23187) followed by 6-dimethylaminopurine. Subsequently, embryos were cultured in NCSU-23 medium supplemented with 4 mg mL(-1) bovine serum albumin for 7 days. In five replicates, 93.0 +/- 7.0% (mean +/- s.e.m.) of attempted reconstructed embryos fused and survived activation (31/31, 15/23, 28/28, 37/37 and 28/28). On Day 7 after activation, the respective blastocyst rates (per successfully reconstructed embryos) were 6% (2/31), 7% (1/15), 7% (2/28), 3% (1/37) and 7% (2/28), resulting in an average of 6.0 +/- 0.8%. Enhanced green fluorescent protein was expressed in all cells of all eight developing blastocysts. Efforts are now directed towards the production of offspring from such transgenic NT blastocysts.


Subject(s)
Animals, Genetically Modified , Blastocyst , Cloning, Organism/methods , Nuclear Transfer Techniques , Swine , Animals , Blastocyst/chemistry , Female , Fibroblasts/chemistry , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Oocytes , Swine/genetics
10.
Am J Physiol Renal Physiol ; 281(3): F546-56, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11502603

ABSTRACT

The vasopressin-induced trafficking of aquaporin-2 (AQP2) water channels in kidney collecting duct is likely mediated by vesicle-targeting proteins (N-ethylmaleimide-sensitive factor attachment protein receptors). Hrs-2 is an ATPase believed to have a modulatory role in regulated exocytosis. To examine whether Hrs-2 is expressed in rat kidney, we carried out RT-PCR combined with DNA sequence analysis and Northern blotting using a digoxigenin-labeled Hrs-2 RNA probe. RT-PCR and Northern blotting revealed that Hrs-2 mRNA is localized in all zones of rat kidney. The presence of Hrs-2 protein in rat kidney was confirmed by immunoblotting, revealing a 115-kDa protein in kidney and brain membrane fractions corresponding to the expected molecular size of Hrs-2. Immunostaining and confocal laser scanning microscopy of LLC-PK(1) cells (a porcine proximal tubule cell line) transfected with Hrs-2 DNA confirmed the specificity of the antibody and revealed that Hrs-2 is mainly localized in intracellular compartments, including cathepsin D-containing lysosomal/endosomal compartments. The cellular and subcellular localization of Hrs-2 in rat kidney was examined by immunocytochemistry and confocal laser scanning microscopy. Hrs-2 immunoreactivity was observed in collecting duct principal cells, and weaker labeling was detected in other nephron segments. The labeling was predominantly present in intracellular vesicles, but labeling was also observed in the apical plasma membrane domains of some cells. Colabeling with AQP2 revealed colocalization in vesicles and apical plasma membrane domains, suggesting a role for Hrs-2 in regulated AQP2 trafficking.


Subject(s)
Adenosine Triphosphatases/genetics , Aquaporins/genetics , Kidney Tubules, Collecting/physiology , Kidney/physiology , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Phosphoproteins , Adenosine Triphosphatases/analysis , Animals , Aquaporin 2 , Aquaporin 6 , Aquaporins/analysis , Cathepsin D/analysis , Cerebellum/cytology , Cerebellum/physiology , Endosomal Sorting Complexes Required for Transport , Endosomes/physiology , Endosomes/ultrastructure , Kidney/cytology , Kidney Tubules, Collecting/cytology , Lysosomes/physiology , Lysosomes/ultrastructure , Membrane Proteins/analysis , Nerve Tissue Proteins/analysis , Organ Specificity , RNA, Messenger/analysis , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Synaptosomal-Associated Protein 25
11.
Hum Mutat ; 18(3): 169-89, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11524729

ABSTRACT

Mutation analysis of metabolic disorders, such as the fatty acid oxidation defects, offers an additional, and often superior, tool for specific diagnosis compared to traditional enzymatic assays. With the advancement of the structural part of the Human Genome Project and the creation of mutation databases, procedures for convenient and reliable genetic analyses are being developed. The most straightforward application of mutation analysis is to specific diagnoses in suspected patients, particularly in the context of family studies and for prenatal/preimplantation analysis. In addition, from these practical uses emerges the possibility to study genotype-phenotype relationships and investigate the molecular pathogenesis resulting from specific mutations or groups of mutations. In the present review we summarize current knowledge regarding genotype-phenotype relationships in three disorders of mitochondrial fatty acid oxidation: very-long chain acyl-CoA dehydrogenase (VLCAD, also ACADVL), medium-chain acyl-CoA dehydrogenase (MCAD, also ACADM), and short-chain acyl-CoA dehydrogenase (SCAD, also ACADS) deficiencies. On the basis of this knowledge we discuss current understanding of the structural implications of mutation type, as well as the modulating effect of the mitochondrial protein quality control systems, composed of molecular chaperones and intracellular proteases. We propose that the unraveling of the genetic and cellular determinants of the modulating effects of protein quality control systems may help to assess the balance between genetic and environmental factors in the clinical expression of a given mutation. The realization that the effect of the monogene, such as disease-causing mutations in the VLCAD, MCAD, and SCAD genes, may be modified by variations in other genes presages the need for profile analyses of additional genetic variations. The rapid development of mutation detection systems, such as the chip technologies, makes such profile analyses feasible. However, it remains to be seen to what extent mutation analysis will be used for diagnosis of fatty acid oxidation defects and other metabolic disorders.


Subject(s)
Fatty Acids/metabolism , Lipid Metabolism, Inborn Errors/genetics , Mitochondria/metabolism , Acyl-CoA Dehydrogenase , Acyl-CoA Dehydrogenase, Long-Chain , Acyl-CoA Dehydrogenases/deficiency , Acyl-CoA Dehydrogenases/genetics , Genotype , Humans , Lipid Metabolism, Inborn Errors/enzymology , Lipid Metabolism, Inborn Errors/metabolism , Mitochondria/enzymology , Mutation , Oxidation-Reduction , Phenotype
12.
Biochem Biophys Res Commun ; 285(2): 320-7, 2001 Jul 13.
Article in English | MEDLINE | ID: mdl-11444845

ABSTRACT

SNAP-23 has an important role in protein-trafficking processes in mammalian cells and until yet two isoforms of SNAP-23 (SNAP-23a and SNAP-23b) have been described. In the present report, we have identified the existence of three new SNAP-23 isoforms (named SNAP-23c, SNAP-23d, and SNAP-23e), which arise from alternative splicing. By RT-PCR all five splice variants were shown to be expressed in four different human inflammatory cells (eosinophils, basophils, neutrophils, and peripheral blood mononuclear cells). Transfection of the human basophilic KU-812 cell line with plasmid constructs containing the cDNAs of the five splice variants located SNAP-23a and SNAP-23b primarily in the plasma membrane. The other three splice variants were localized both intracellularly and in the plasma membrane.


Subject(s)
Alternative Splicing , Carrier Proteins/genetics , Genetic Variation , Granulocytes/metabolism , Leukocytes, Mononuclear/metabolism , Amino Acid Sequence , Animals , Base Sequence , Basophils/metabolism , Carrier Proteins/chemistry , Cell Line , Eosinophils/metabolism , Exons , Humans , Introns , Mammals , Molecular Sequence Data , Neutrophils/metabolism , Protein Isoforms/chemistry , Protein Isoforms/genetics , Qb-SNARE Proteins , Qc-SNARE Proteins , Recombinant Fusion Proteins/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Sequence Homology, Amino Acid , Transcription, Genetic , Transfection
13.
J Inherit Metab Dis ; 24(2): 189-212, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11405340

ABSTRACT

Molecular chaperones are present in the various compartments of the cell and assist the folding of newly synthesized proteins. Compared to wild-type proteins, missense mutant proteins are generally synthesized in a normal fashion, but may be impaired in their folding. A broad array of diseases that are due to misfolding of mutant proteins may be labelled conformational diseases: aggregation diseases, such as Alzheimer disease; diseases caused by negative dominance from misfolded structural proteins, such as hypertrophic cardiomyopathy; and disorders where the misfolded protein is degraded by intracellular proteases. Many metabolic disorders belong to this last category, where the so-called protein quality control systems, comprising chaperones and proteases, attempt to eliminate folding intermediates or misfolded proteins. On the basis of in vitro experiments with a limited number of missense mutations identified in patients with phenylalanine hydroxylase and fatty acid oxidation deficiencies, we discuss the cellular fate of missense mutant proteins. We find that the balance between folding to functional conformers, retention (holding) and degradation of folding intermediates or misfolded proteins is dependent on the nature of the mutation and on the efficiency of the quality control. For example, low temperature may promote formation of functional conformers, while elevated temperature usually promotes retention and degradation. We conclude that disorders caused by many missense mutations are complex diseases in which the mutation itself is a necessary major primary component, but that its effect may be modified by cellular conditions and possibly by genetic variations in the quality control systems. We suggest that this new knowledge about cell handling may open new avenues of understanding of the cell pathology and treatment of patients with metabolic disorders.


Subject(s)
Metabolism, Inborn Errors/genetics , Molecular Chaperones/genetics , Protein Folding , Animals , Humans , Protein Conformation
14.
Eur J Hum Genet ; 9(11): 815-22, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11781697

ABSTRACT

The function of a series of LDL receptor GFP fusion proteins with different, flexible, unstructured spacer regions was analysed. An optimised version of the fusion protein was used to analyse the effect of an LDL receptor mutation (W556S) found in FH patients and characterised as transport defective. In cultured liver cells this mutation was found to inhibit the transport of LDL receptor GFP fusion protein to the cell surface, thus leading to impaired internalisation of fluorescent labelled LDL. Co-localisation studies confirmed the retention of the mutant protein in the endoplasmic reticulum. Wild type (WT) and W556S LDL receptor GFP fusion proteins were expressed in mouse liver by means of hydrodynamic delivery of naked DNA. Two days after injection liver samples were analysed for GFP fluorescence. The WT LDL receptor GFP protein was located on the cell surface whereas the W556S LDL receptor GFP protein was retained in intracellular compartments. Thus, the GFP-tagged LDL receptor protein allows both detailed time lapse analysis and evaluations in animals for the physiological modelling of mutations. This method should be generally applicable in functional testing of gene products for aberrant processing.


Subject(s)
Receptors, LDL/physiology , Animals , Biological Transport , Cell Line , Endocytosis , Genotype , Green Fluorescent Proteins , Humans , Hyperlipoproteinemia Type II/genetics , Lipoproteins, LDL/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Inbred Strains , Mice, Knockout , Microscopy, Confocal , Mutation , Receptors, LDL/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transfection
15.
Am J Hum Genet ; 67(5): 1095-103, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11013134

ABSTRACT

Acyl-CoA dehydrogenase (ACAD) defects in isoleucine and valine catabolism have been proposed in clinically diverse patients with an abnormal pattern of metabolites in their urine, but they have not been proved enzymatically or genetically, and it is unknown whether one or two ACADs are involved. We investigated a patient with isolated 2-methylbutyrylglycinuria, suggestive of a defect in isoleucine catabolism. Enzyme assay of the patient's fibroblasts, using 2-methylbutyryl-CoA as substrate, confirmed the defect. Sequence analysis of candidate ACADs revealed heterozygosity for the common short-chain ACAD A625 variant allele and no mutations in ACAD-8 but a 100-bp deletion in short/branched-chain ACAD (SBCAD) cDNA from the patient. Our identification of the SBCAD gene structure (11 exons; >20 kb) enabled analysis of genomic DNA. This showed that the deletion was caused by skipping of exon 10, because of homozygosity for a 1228G-->A mutation in the patient. This mutation was not present in 118 control chromosomes. In vitro transcription/translation experiments and overexpression in COS cells confirmed the disease-causing nature of the mutant SBCAD protein and showed that ACAD-8 is an isobutyryl-CoA dehydrogenase and that both wild-type proteins are imported into mitochondria and form tetramers. In conclusion, we report the first mutation in the SBCAD gene, show that it results in an isolated defect in isoleucine catabolism, and indicate that ACAD-8 is a mitochondrial enzyme that functions in valine catabolism.


Subject(s)
Amino Acid Metabolism, Inborn Errors/enzymology , Amino Acid Metabolism, Inborn Errors/genetics , Isoleucine/metabolism , Oxidoreductases Acting on CH-CH Group Donors , Oxidoreductases/deficiency , Valine/metabolism , Alternative Splicing/genetics , Amino Acid Metabolism, Inborn Errors/metabolism , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Child, Preschool , Consanguinity , DNA Mutational Analysis , Enzyme Stability , Exons , Female , Heterozygote , Humans , Introns , Male , Mitochondria/enzymology , Mitochondria/metabolism , Mutation/genetics , Oxidoreductases/genetics , Oxidoreductases/metabolism , Pakistan , Protein Transport , Sequence Deletion/genetics , Transfection
16.
Mamm Genome ; 11(10): 899-905, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11003706

ABSTRACT

We have determined the cDNA sequence and exon/intron structure of the human CLPX gene encoding a human ortholog of the E. coli ClpX chaperone and protease subunit. The CLPX gene comprises 14 exons and encodes a 633-amino acid-long precursor polypeptide. The polypeptide contains an N-terminal putative mitochondrial transit peptide, and expression of a full-length ClpX cDNA tagged at its C-terminus (Myc-His) shows that the polypeptide is transported into mitochondria. FISH analysis localized the CLPX gene to human Chromosome (Chr) 15q22.1-22.32. This localization was refined by radiation hybrid mapping placing the CLPX gene 4.6 cR distal to D15S159. Murine ClpX cDNA was sequenced, and the mouse Clpx locus was mapped to a position between 31 and 42 cM offset from the centromere on mouse Chr 9. Experimental observations indicate the presence of a pseudogene in the mouse genome and sequence variability between mouse ClpX cDNAs from different strains. Alignment of the human and mouse ClpX amino acid sequences with ClpX sequences from other organisms shows that they display the typical modular organization of domains with one AAA(+) domain common to a large group of ATPases and several other domains conserved in ClpX orthologs linked by non-conserved sequences. Notably, a C-4 zinc finger type motif is recognized in human and mouse ClpX. This motif of so far unknown function is present only in a subset of the known ClpX sequences.


Subject(s)
Adenosine Triphosphatases/genetics , Chromosomes, Human, Pair 15 , Mitochondria/genetics , ATPases Associated with Diverse Cellular Activities , Adenosine Triphosphatases/physiology , Amino Acid Sequence , Animals , Chromosome Mapping , DNA/analysis , Endopeptidase Clp , Escherichia coli Proteins , Gene Expression , Genome , Humans , Mice , Mice, Inbred C57BL , Molecular Chaperones , Molecular Sequence Data , Protein Conformation , Sequence Homology, Amino Acid , Subcellular Fractions
17.
J Inherit Metab Dis ; 23(5): 441-7, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10947197

ABSTRACT

Many disease-causing point mutations do not seriously compromise synthesis of the affected polypeptide but rather exert their effects by impairing subsequent protein folding or stability of the folded protein. This often results in rapid degradation of the affected protein. The concepts of such 'conformational disease' are illustrated by reference to cystic fibrosis, phenylketonuria and short-chain acyl-CoA dehydrogenase deficiency. Other cellular components such as chaperones and proteases, as well as environmental factors, may combine to modulate the phenotype of such disorders and this may open up new therapeutic approaches.


Subject(s)
Genetic Diseases, Inborn/metabolism , Proteins/chemistry , Proteins/metabolism , Acyl-CoA Dehydrogenase , Acyl-CoA Dehydrogenases/deficiency , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cytosol/metabolism , Endoplasmic Reticulum/metabolism , Genetic Diseases, Inborn/genetics , Humans , Mitochondria/metabolism , Mutation , Phenylketonurias/genetics , Phenylketonurias/metabolism , Protein Conformation , Protein Folding , Proteins/genetics
18.
J Biol Chem ; 275(43): 33861-8, 2000 Oct 27.
Article in English | MEDLINE | ID: mdl-10906332

ABSTRACT

The low density lipoprotein (LDL) receptor is responsible for removing the majority of the LDL cholesterol from the plasma. Mutations in the LDL receptor gene cause the disease familial hypercholesterolemia (FH). Approximately 50% of the mutations in the LDL receptor gene in patients with FH lead to receptor proteins that are retained in the endoplasmic reticulum (ER). Misfolding of mutant LDL receptors is a probable cause of this ER retention, resulting in no functional LDL receptors at the cell surface. However, the specific factors and mechanisms responsible for retention of mutant LDL receptors are unknown. In the present study we show that the molecular chaperone Grp78/BiP co-immunoprecipitates with both the wild type and two different mutant (W556S and C646Y) LDL receptors in lysates obtained from human liver cells overexpressing wild type or mutant LDL receptors. A pulse-chase study shows that the interaction between the wild type LDL receptor and Grp78 is no longer detectable after 2(1/2) h, whereas it persists for more than 4 h with the mutant receptors. Furthermore, about five times more Grp78 is co-immunoprecipitated with the mutant receptors than with the wild type receptor suggesting that Grp78 is involved in retention of mutant LDL receptors in the ER. Overexpression of Grp78 causes no major alterations on the steady state level of active LDL receptors at the cell surface. However, overexpression of Grp78 decreases the processing rate of newly synthesized wild type LDL receptors. This indicates that the Grp78 interaction is a rate-limiting step in the maturation of the wild type LDL receptor and that Grp78 may be an important factor in the quality control of newly synthesized LDL receptors.


Subject(s)
Endoplasmic Reticulum/metabolism , HSP70 Heat-Shock Proteins/physiology , Membrane Proteins/physiology , Receptors, LDL/metabolism , 6-Aminonicotinamide/pharmacology , Amino Acid Sequence , Cells, Cultured , Endoplasmic Reticulum Chaperone BiP , Humans , Molecular Sequence Data , Molecular Weight , Mutation
19.
Mamm Genome ; 11(4): 275-80, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10754102

ABSTRACT

Mutations that cause accumulation or rapid degradation owing to protein misfolding are a frequent cause of inherited disease in humans. In Escherichia coli, Clpp protease is one of the components of the protein quality control system that handles misfolded proteins. In the present study, we have characterized the mouse Clpp cDNA sequence, the organization of the mouse gene, the chromosomal localization, and the tissue-specific expression pattern. Moreover. the cellular localization and processing of mouse Clpp was studied by overexpression in transfected eukaryotic cells. Our results indicate that mouse and human Clpp have similar roles, and they provide the molecular basis for establishing a Clpp knockout mouse and to study its phenotype, thereby shedding light on a possible role of Clpp in human disease.


Subject(s)
Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Animals , Base Sequence , Chromosome Mapping/veterinary , DNA Primers , DNA, Complementary , Endopeptidase Clp , Exons , Humans , Introns , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
20.
Cell Biol Int ; 23(11): 719-28, 2000.
Article in English | MEDLINE | ID: mdl-10736196

ABSTRACT

The expression and intracellular localization of the Tetrahymena homolog of 4-hydroxyphenylpyruvate dioxygenase (HPPD) were investigated in wild-type Tetrahymena thermophila strain B1868 VII and the mutant strains IIG8, defective in food vacuole formation, MS-1, blocked in secretion of lysosomal enzymes, and SB 281, defective in mucocyst maturation. Immunoelectron microscopy and confocal laser scanning microscopy demonstrated that Tetrahymena HPPD primarily localized to membranes of the endoplasmic reticulum. In addition, Tetrahymena HPPD was detected in association with membranes of the Golgi apparatus, and transport vesicles in exponentially growing wild-type and mutant strains. In starved cells, Tetrahymena HPPD localized exclusively to membranes of small vesicles. Since no de novo synthesis of Tetrahymena HPPD takes place in cells starved for more than 30 min, these results suggest that there is a flow of Tetrahymena HPPD from the endoplasmic reticulum to small vesicles, possibly via the Golgi apparatus, and that Tetrahymena HPPD contains a signal for vesicle membrane retrieval or retention.


Subject(s)
4-Hydroxyphenylpyruvate Dioxygenase/metabolism , Endoplasmic Reticulum/enzymology , Intracellular Membranes/enzymology , Tetrahymena thermophila/enzymology , 4-Hydroxyphenylpyruvate Dioxygenase/genetics , Animals , Bacteria/enzymology , Endoplasmic Reticulum/ultrastructure , Intracellular Membranes/ultrastructure , Mammals , Recombinant Fusion Proteins/metabolism , Tetrahymena thermophila/genetics , Vacuoles/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...