Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Biochim Biophys Acta Mol Basis Dis ; 1869(8): 166808, 2023 12.
Article in English | MEDLINE | ID: mdl-37454773

ABSTRACT

Ionic calcium (Ca2+) is a key messenger in signal transduction and its mitochondrial uptake plays an important role in cell physiology. This uptake is mediated by the mitochondrial Ca2+ uniporter (MCU), which is regulated by EMRE (essential MCU regulator) encoded by the SMDT1 (single-pass membrane protein with aspartate rich tail 1) gene. This work presents the genetic, clinical and cellular characterization of two patients harbouring SMDT1 variants and presenting with muscle problems. Analysis of patient fibroblasts and complementation experiments demonstrated that these variants lead to absence of EMRE protein, induce MCU subcomplex formation and impair mitochondrial Ca2+ uptake. However, the activity of oxidative phosphorylation enzymes, mitochondrial morphology and membrane potential, as well as routine/ATP-linked respiration were not affected. We hypothesize that the muscle-related symptoms in the SMDT1 patients result from aberrant mitochondrial Ca2+ uptake.


Subject(s)
Calcium Channels , Calcium , Humans , Calcium/metabolism , Calcium Channels/genetics , Calcium Channels/metabolism , Ion Transport , Mitochondria/genetics , Mitochondria/metabolism , Muscles/metabolism
2.
J Inherit Metab Dis ; 46(1): 43-54, 2023 01.
Article in English | MEDLINE | ID: mdl-36117148

ABSTRACT

Infantile nephropathic cystinosis (INC) is an inheritable lysosomal storage disorder characterized by lysosomal cystine accumulation, progressive kidney disease, and multiple extrarenal complications (ERCs). Cysteamine postpones the onset of end-stage kidney disease (ESKD) and reduces the incidence of ERCs; however, cysteamine is generally initiated upon establishment of the renal Fanconi syndrome (FS) and partial loss of kidney function, whereas data on long-term effects of cysteamine administered from neonatal age are lacking. An international multicenter retrospective cohort study of siblings with INC was set up to investigate the outcome in relation to age at initiation of cysteamine versus CTNS genotype, with attention to patients treated with cysteamine from neonatal age. None of the siblings treated from neonatal age (n = 9; age 10 ± 6 years) had reached ESKD, while 22% of their index counterparts (n = 9; age 14 ± 5 years) had commenced renal replacement therapy. Siblings treated with cysteamine from the onset of symptoms at a younger age compared with their index counterparts, reached ESKD at a significant older age (13 ± 3 vs. 10 ± 3 years, p = 0.002). In contrast, no significant difference in ERCs was observed between sibling and index patients, independently from the age at initiation of cysteamine. The CTNS genotype had no impact on the overall outcome in this cohort. In INC, presymptomatic treatment with cysteamine results in a better renal outcome in comparison to treatment initiated from the onset of symptoms. This justifies including cystinosis into newborn screening programs. SYNOPSIS: In infantile nephropathic cystinosis, presymptomatic treatment with cysteamine improves the renal outcome which justifies the inclusion of cystinosis into newborn screening programs.


Subject(s)
Cystinosis , Fanconi Syndrome , Kidney Failure, Chronic , Infant, Newborn , Humans , Child, Preschool , Child , Adolescent , Young Adult , Adult , Cystinosis/drug therapy , Cystinosis/genetics , Cystinosis/complications , Cysteamine/therapeutic use , Siblings , Cohort Studies , Retrospective Studies , Fanconi Syndrome/drug therapy , Fanconi Syndrome/genetics , Kidney Failure, Chronic/etiology
3.
Methods Mol Biol ; 2227: 83-96, 2021.
Article in English | MEDLINE | ID: mdl-33847933

ABSTRACT

The complement system is a key part of innate immunity. However, if the system becomes dysregulated, damage to healthy host cells can occur, especially to the glomerular cells of the kidney. The convertases of the alternative pathway of the complement system play a crucial role in complement activation. In healthy conditions, their activity is strictly regulated. In patients with diseases caused by complement alternative pathway dysregulation, such as C3 glomerulopathy and atypical hemolytic uremic syndrome, factors can be present in the blood that disturb this delicate balance, leading to convertase overactivity. Such factors include C3 nephritic factors, which are autoantibodies against the C3 convertase that prolong its activity, or genetic variants resulting in a stabilized convertase complex. This chapter describes a method in which the activity and stability of the alternative pathway convertases can be measured to detect aberrant serum factors causing convertase overactivity.


Subject(s)
Complement C3-C5 Convertases/metabolism , Complement Hemolytic Activity Assay/methods , Complement Pathway, Alternative , Animals , Atypical Hemolytic Uremic Syndrome/blood , Atypical Hemolytic Uremic Syndrome/diagnosis , Atypical Hemolytic Uremic Syndrome/immunology , Complement Activation , Complement C3/immunology , Complement C3 Nephritic Factor/analysis , Complement C3 Nephritic Factor/immunology , Complement C3-C5 Convertases/analysis , Complement Pathway, Alternative/immunology , Glomerulonephritis/blood , Glomerulonephritis/diagnosis , Glomerulonephritis/immunology , Guinea Pigs , Humans , Rabbits
5.
Nephron ; 141(2): 133-146, 2019.
Article in English | MEDLINE | ID: mdl-30554218

ABSTRACT

Mutations in the CTNS gene encoding the lysosomal membrane cystine transporter cystinosin are the cause of cystinosis, an autosomal recessive lysosomal storage disease. More than 140 CTNS mutations have been reported worldwide. Recent studies have discovered that cystinosin exerts other key cellular functions beyond cystine transport such as regulation of oxidative state, lysosomal dynamics and autophagy. Here, we review the different mutations described in the CTNS gene and the geographical distribution of incidence. In addition, the characteristics of the various mutations in relation to the functions of cystinosin needs to be further elucidated. In this review, we highlight the functional consequences of the different mutations in correlation with the clinical phenotypes. Moreover, we propose how this understanding would be fundamental for the development of new technologies through targeted gene therapy, holding promises for a possible cure of the kidney and extra-renal phenotypes of cystinosis.


Subject(s)
Amino Acid Transport Systems, Neutral/genetics , Cystinosis/genetics , DNA Repair , Mutation , Animals , Cystinosis/epidemiology , Disease Models, Animal , Genotype , Geography , Humans , Incidence , Phenotype
6.
PLoS One ; 12(8): e0182350, 2017.
Article in English | MEDLINE | ID: mdl-28771540

ABSTRACT

OBJECTIVE: The aim of this study is to evaluate the diagnostic performance of FDG-PET/CT for the detection of residual disease after (chemo)radiotherapy in patients with head and neck squamous cell carcinoma (HNSCC) and to evaluate the prognostic value of the FDG-PET/CT findings. METHODS: Patients with HNSCC who underwent FDG-PET/CT after (chemo)radiotherapy were studied retrospectively. RESULTS: 104 FDG-PET/CT-scans were performed at a median of 13.2 weeks post-treatment (5.4-19.0 weeks). The diagnostic performance was time dependent with decreasing sensitivity and slightly increasing specificity over time. Sensitivity, specificity, PPV and NPV at 9 months after imaging were 91%, 87%, 77% and 95%, respectively. In a logistic regression model, the odds of a correct FDG-PET/CT increased with 33% every additional week after end of therapy (p = 0.01) and accuracy plateaued after 11 weeks (97%; p<0.001). A complete response on FDG-PET/CT was associated with an overall survival benefit (50.7 versus 10.3 months; p<0.001). Residual disease on FDG-PET/CT increased the risk of death 8-fold (p<0.001). CONCLUSION: FDG-PET/CT is able to detect residual disease after (chemo)radiotherapy, with an optimal time point for scanning between 11-12 weeks after therapy. However, a reevaluation is probably necessary 10-12 months after the FDG-PET/CT to detect late recurrences. In addition, FDG-PET/CT can guide decisions about neck dissection and identifies patients with poor prognosis.


Subject(s)
Carcinoma, Squamous Cell/diagnosis , Head and Neck Neoplasms/diagnosis , Positron Emission Tomography Computed Tomography , Radiopharmaceuticals/chemistry , Adult , Aged , Aged, 80 and over , Area Under Curve , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/radiotherapy , Female , Fluorodeoxyglucose F18/chemistry , Head and Neck Neoplasms/mortality , Head and Neck Neoplasms/radiotherapy , Humans , Kaplan-Meier Estimate , Logistic Models , Male , Middle Aged , Neoplasm, Residual , Prognosis , Proportional Hazards Models , ROC Curve , Retrospective Studies , Sensitivity and Specificity , Squamous Cell Carcinoma of Head and Neck
7.
Am J Hematol ; 91(12): E482-E490, 2016 12.
Article in English | MEDLINE | ID: mdl-27643674

ABSTRACT

TMPRSS6 variants that affect protein function result in impaired matriptase-2 function and consequently uninhibited hepcidin production, leading to iron refractory iron deficiency anemia (IRIDA). This disease is characterized by microcytic, hypochromic anemia and serum hepcidin values that are inappropriately high for body iron levels. Much is still unknown about its pathophysiology, genotype-phenotype correlation, and optimal clinical management. We describe 14 different TMPRSS6 variants, of which 9 are novel, in 21 phenotypically affected IRIDA patients from 20 families living in the Netherlands; 16 out of 21 patients were female. In 7 out of 21 cases DNA sequencing and multiplex ligation dependent probe amplification demonstrated only heterozygous TMPRSS6 variants. The age at presentation, disease severity, and response to iron supplementation were highly variable, even for patients and relatives with similar TMPRSS6 genotypes. Mono-allelic IRIDA patients had a milder phenotype with respect to hemoglobin and MCV and presented significantly later in life with anemia than bi-allelic patients. Transferrin saturation (TSAT)/hepcidin ratios were lower in IRIDA probands than in healthy relatives. Most patients required parenteral iron. Genotype alone was not predictive for the response to oral iron. We conclude that IRIDA is a genotypically and phenotypically heterogeneous disease. The high proportion of female patients and the discrepancy between phenotypes of probands and relatives with the same genotype, suggest a complex interplay between genetic and acquired factors in the pathogenesis of IRIDA. In the absence of inflammation, the TSAT/hepcidin ratio is a promising diagnostic tool, even after iron supplementation has been given. Am. J. Hematol. 91:E482-E490, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Anemia, Iron-Deficiency , Adolescent , Adult , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Middle Aged , Netherlands , Young Adult
8.
BMJ Case Rep ; 20162016 Feb 15.
Article in English | MEDLINE | ID: mdl-26880823

ABSTRACT

We present the case of a 18-month-old girl with renal and cardiac manifestations of atypical haemolytic uraemic syndrome (aHUS), and a novel complement factor H mutation. Transient haematological remission was achieved with intensive plasmapheresis, but cardiac function deteriorated and renal function was not restored. Initiation of eculizumab after 6 months of dialysis significantly improved organ function. At 43 months after presentation, haematological values had normalised and cardiac function had improved. Dialysis was discontinued after 10 months (the longest reported time in a patient with aHUS) and the estimated glomerular filtration rate had recovered to 70 mL/min/1.73 m(2). In conclusion, treatment of aHUS with eculizumab, even after long-term dialysis, can significantly improve renal function. Discontinuation of dialysis and resolution of cardiac function has implications on the potential recovery and treatment choice of such patients. Earlier initiation of eculizumab, however, might have prevented the irreversible renal sclerosis and cardiac dysfunction.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Atypical Hemolytic Uremic Syndrome/drug therapy , Cardiomyopathies/etiology , Renal Insufficiency/etiology , Atypical Hemolytic Uremic Syndrome/complications , Atypical Hemolytic Uremic Syndrome/diagnosis , Cardiomyopathies/therapy , Diagnosis, Differential , Female , Humans , Infant , Kidney/pathology , Plasmapheresis , Renal Insufficiency/pathology , Renal Insufficiency/therapy
9.
Minerva Chir ; 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26771252

ABSTRACT

BACKGROUND: The aim of this study is to investigate the role of Octreotide LAR in secondary prevention in patients with chemotherapy-induced diarrhea. METHODS: In this study, patients experiencing CID ≥ grade 2 received 30 mg long-acting octreotide as a monthly injection and the next chemotherapy dose was administrated with a 25% dose decrease. If no CID ≥ grade 2 occurred, subsequent chemotherapy doses were increased to the initial 100% values. The primary endpoint of the study was the diarrhea control rate (< grade 2) for patients receiving the optimal dose of chemotherapy for a minimum of 2 cycles. RESULTS: Twenty-nine patients were included. Ten patients experienced no improvement or ended the study very early after the first injection of octreotide LAR. Nineteen patients had a reduction in the grade of diarrhea after the first administration of Octreotide LAR and a reduced chemotherapy dose. Seven of them (24%) did not reach the end of the study because of disease progression (6) or lost in follow-up (1). Ultimately 12 patients (41%) continued the study till the end. In ten of these twelve patients, there was a significant and persisting reduction of diarrhea while receiving full dose chemotherapy. CONCLUSION: This study suggests that monthly injections with long-acting octreotide might be used as a secondary prevention of chemotherapy-induced diarrhea. Its usefulness and optimal dosage in secondary prevention in combination with antidiarrheal agents needs further research.

10.
Pediatr Nephrol ; 31(8): 1267-79, 2016 08.
Article in English | MEDLINE | ID: mdl-26267676

ABSTRACT

Nephrotic syndrome (NS) is a common kidney disease associated with a significantly increased risk of thrombotic events. Alterations in plasma levels of pro- and anti-coagulant factors are involved in the pathophysiology of venous thrombosis in NS. However, the fact that the risk of both venous and arterial thrombosis is elevated in NS points to an additional role for blood platelets. Increased platelet counts and platelet hyperactivity have been observed in nephrotic children. Platelet hyperaggregability, increased release of active substances, and elevated surface expression of activation-dependent platelet markers have been documented. The mechanisms underlying those platelet alterations are multifactorial and are probably due to changes in plasma levels of platelet-interfering proteins and lipid changes, as a consequence of nephrosis. The causal relationship between platelet alterations seen in NS and the occurrence of thromboembolic phenomena remains unclear. Moreover, the efficiency of prophylactic treatment using antiplatelet agents for the prevention of thrombotic complications in nephrotic patients is also unknown. Thus, antiplatelet medication is currently not generally recommended for routine prophylactic therapy.


Subject(s)
Blood Platelets/pathology , Nephrotic Syndrome/blood , Child , Humans , Nephrotic Syndrome/physiopathology
11.
Eur J Hum Genet ; 22(7): 888-95, 2014 Jul.
Article in English | MEDLINE | ID: mdl-23963297

ABSTRACT

Patients with cutis laxa (CL) have wrinkled, sagging skin with decreased elasticity. Skin symptoms are associated with variable systemic involvement. The most common, genetically highly heterogeneous form of autosomal recessive CL, ARCL2, is frequently associated with variable metabolic and neurological symptoms. Progeroid symptoms, dysmorphic features, hypotonia and psychomotor retardation are highly overlapping in the early phase of these disorders. This makes the genetic diagnosis often challenging. In search for discriminatory symptoms, we prospectively evaluated clinical, neurologic, metabolic and genetic features in our patient cohort referred for suspected ARCL. From a cohort of 26 children, we confirmed mutations in genes associated with ARCL in 16 children (14 probands), including 12 novel mutations. Abnormal glycosylation and gyration abnormalities were mostly, but not always associated with ATP6V0A2 mutations. Epilepsy was most common in ATP6V0A2 defects. Corpus callosum dysgenesis was associated with PYCR1 and ALDH18A1 mutations. Dystonic posturing was discriminatory for PYCR1 and ALDH18A1 defects. Metabolic markers of mitochondrial dysfunction were found in one patient with PYCR1 mutations. So far unreported white matter abnormalities were found associated with GORAB and RIN2 mutations. We describe a large cohort of CL patients with neurologic involvement. Migration defects and corpus callosum hypoplasia were not always diagnostic for a specific genetic defect in CL. All patients with ATP6V0A2 defects had abnormal glycosylation. To conclude, central nervous system and metabolic abnormalities were discriminatory in this genetically heterogeneous group, although not always diagnostic for a certain genetic defect in CL.


Subject(s)
Agenesis of Corpus Callosum , Cutis Laxa , Epilepsy , Adolescent , Agenesis of Corpus Callosum/diagnosis , Agenesis of Corpus Callosum/genetics , Agenesis of Corpus Callosum/metabolism , Agenesis of Corpus Callosum/pathology , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Child , Child, Preschool , Cutis Laxa/diagnosis , Cutis Laxa/genetics , Cutis Laxa/metabolism , Cutis Laxa/pathology , Epilepsy/diagnosis , Epilepsy/genetics , Epilepsy/metabolism , Epilepsy/pathology , Female , Glycosylation , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Humans , Male , Mitochondrial Proton-Translocating ATPases/genetics , Mitochondrial Proton-Translocating ATPases/metabolism , Prospective Studies , Pyrroline Carboxylate Reductases/genetics , Pyrroline Carboxylate Reductases/metabolism , delta-1-Pyrroline-5-Carboxylate Reductase
12.
Hum Mutat ; 34(12): 1721-6, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24123792

ABSTRACT

The advent of massive parallel sequencing is rapidly changing the strategies employed for the genetic diagnosis and research of rare diseases that involve a large number of genes. So far it is not clear whether these approaches perform significantly better than conventional single gene testing as requested by clinicians. The current yield of this traditional diagnostic approach depends on a complex of factors that include gene-specific phenotype traits, and the relative frequency of the involvement of specific genes. To gauge the impact of the paradigm shift that is occurring in molecular diagnostics, we assessed traditional Sanger-based sequencing (in 2011) and exome sequencing followed by targeted bioinformatics analysis (in 2012) for five different conditions that are highly heterogeneous, and for which our center provides molecular diagnosis. We find that exome sequencing has a much higher diagnostic yield than Sanger sequencing for deafness, blindness, mitochondrial disease, and movement disorders. For microsatellite-stable colorectal cancer, this was low under both strategies. Even if all genes that could have been ordered by physicians had been tested, the larger number of genes captured by the exome would still have led to a clearly superior diagnostic yield at a fraction of the cost.


Subject(s)
Exome , Genetic Diseases, Inborn/diagnosis , Genetic Diseases, Inborn/genetics , High-Throughput Nucleotide Sequencing , Sequence Analysis, DNA , Genetic Counseling , Genetic Testing , High-Throughput Nucleotide Sequencing/methods , High-Throughput Nucleotide Sequencing/standards , Humans , Sequence Analysis, DNA/methods , Sequence Analysis, DNA/standards
13.
Int J Pharm ; 452(1-2): 14-35, 2013 Aug 16.
Article in English | MEDLINE | ID: mdl-23711732

ABSTRACT

Since their introduction circa 35 years ago, calcineurin-inhibitors (CNI) have become the cornerstone of immunosuppressive therapy in solid organ transplantation. However, CNI's possess a narrow therapeutic index with potential severe consequences of drug under- or overexposure. This demands a meticulous policy of Therapeutic Drug Monitoring (TDM) to optimize outcome. In clinical practice optimal dosing is difficult to achieve due to important inter- and intraindividual variation in CNI pharmacokinetics. A complex and often interdependent set of factors appears relevant in determining drug exposure. These include recipient characteristics such as age, race, body composition, organ function, and food intake, but also graft-related characteristics such as: size, donor-age, and time after transplantation can be important. Fundamental (in vitro) and clinical studies have pointed out the intrinsic relation between the aforementioned variables and the functional capacity of enzymes and transporters involved in CNI metabolism, primarily located in intestine, liver and kidney. Commonly occurring polymorphisms in genes responsible for CNI metabolism (CYP3A4, CYP3A5, CYP3A7, PXR, POR, ABCB1 (P-gp) and possibly UGT) are able to explain an important part of interindividual variability. In particular, a highly prevalent SNP in CYP3A5 has proven to be an important determinant of CNI dose requirements and drug-dose-interactions. In addition, a discrepancy in genotype between graft and receptor has to be taken into account. Furthermore, common phenomena in solid organ transplantation such as inflammation, ischemia- reperfusion injury, graft function, co-medication, altered food intake and intestinal motility can have a differential effect on the expression enzymes and transporters involved in CNI metabolism. Notwithstanding the built-up knowledge, predicting individual CNI pharmacokinetics and dose requirements on the basis of current clinical and experimental data remains a challenge.


Subject(s)
Calcineurin Inhibitors , Immunosuppressive Agents/pharmacokinetics , Organ Transplantation , Animals , Cyclosporine/pharmacokinetics , Humans , Intestinal Mucosa/metabolism , Kidney/metabolism , Liver/metabolism , Tacrolimus/pharmacokinetics
14.
Pediatr Neurol ; 42(3): 227-30, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20159436

ABSTRACT

Mutations in the SCO2 gene [SCO cytochrome oxidase deficient homolog 2 (yeast)] causing cytochrome c oxidase deficiency have been reported in at least in 26 patients with fatal infantile cardioencephalomyopathy. Mutation 1541G > A affecting protein stability is associated with the majority of cases, and the other 11 described mutations have more serious deleterious structural consequences for the protein product. Reported here is a novel case caused by compound heterozygosity of SCO2. The child presented at the age of 3 weeks with failure-to-thrive, muscular hypotonia, hypertrophic cardiomyopathy, and lactic acidemia. Leigh syndrome was diagnosed based on magnetic resonance imaging findings. Immunohistochemical and enzymatic investigations on muscle indicated totally absent cytochrome c oxidase activity. Both parents had mild mental retardation. Sequence analysis in the patient and in his parents revealed heterozygous mutation c.418G > A in exon 2 inherited from the father and maternally inherited heterozygous insertion of 19bp at position 17 in the coding region of the SCO2 gene. Respiratory chain enzyme activity measurements indicated normal activity in both parents, although the mother's cytochrome c oxidase activity was lower. This gene may be involved in the etiology of the mother's mental retardation.


Subject(s)
Brain Damage, Chronic/complications , Brain Damage, Chronic/genetics , Cardiomyopathies/complications , Cardiomyopathies/genetics , Carrier Proteins/genetics , Mitochondrial Proteins/genetics , Point Mutation/genetics , Age Factors , DNA Mutational Analysis , Failure to Thrive/diagnosis , Failure to Thrive/etiology , Humans , Immunohistochemistry , Infant , Infant, Newborn , Male , Molecular Chaperones , Mutagenesis, Insertional/genetics
15.
Clin J Am Soc Nephrol ; 4(10): 1593-600, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19808243

ABSTRACT

BACKGROUND AND OBJECTIVES: Frequently relapsing and steroid-dependent minimal-change nephrotic syndrome (MCNS) that originates in childhood can persist after puberty in >20% of patients. These patients require immunosuppressive treatment during several decades of their life. We examined long-term adverse effects of persistent nephrotic syndrome and immunosuppressive medications, focusing on renal function, growth, obesity, osteoporosis, hypertension, ocular complications, and fertility in adult patients with biopsy-proven childhood-onset MCNS. Molecular analysis was performed to evaluate a possible association of a complicated course of MCNS with podocyte gene mutations. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: We performed a prospective clinical examination of 15 adult patients that included serum and urine analysis; dual-energy x-ray absorptiometry; ophthalmologic examination; semen examination; and molecular analysis of NPHS1, NPHS2, CD2AP, and ACTN4 genes. RESULTS: All patients had normal GFR. Most frequent long-term complications were hypertension (in seven of 15 patients) and osteoporosis in one third of patients. Oligozoospermia was found in one patient, reduced sperm motility in four of eight patients, and teratozoospermia in six of eight patients. Ophthalmologic examination revealed myopia in 10 of 15 patients and cataract in three of 15 patients. CONCLUSIONS: Children with MCNS that persists after puberty are at risk for complications such as osteoporosis, hypertension, cataract, and sperm abnormalities. Our study underscores a need for more effective and less toxic therapies for relapsing MCNS.


Subject(s)
Nephrosis, Lipoid/therapy , Absorptiometry, Photon , Adolescent , Adult , Biopsy , Bone Density , Child , Child, Preschool , Eye Diseases/etiology , Female , Fertility , Humans , Infant , Intracellular Signaling Peptides and Proteins/genetics , Kidney/physiopathology , Male , Membrane Proteins/genetics , Nephrosis, Lipoid/complications , Nephrosis, Lipoid/physiopathology , Recurrence
16.
BMJ Case Rep ; 20092009.
Article in English | MEDLINE | ID: mdl-21686774

ABSTRACT

To identify the biochemical and molecular genetic defect in a 16-year-old patient presenting with apical hypertrophic cardiomyopathy and neuropathy suspected for a mitochondrial disorder.Measurement of the mitochondrial energy-generating system (MEGS) capacity in muscle and enzyme analysis in muscle and fibroblasts were performed. Relevant parts of the mitochondrial DNA were analysed by sequencing.A homoplasmic nonsense mutation m.8529G→A (p.Trp55X) was found in the mitochondrial ATP8 gene in the patient's fibroblasts and muscle tissue. Reduced complex V activity was measured in the patient's fibroblasts and muscle tissue, and was confirmed in cybrid clones containing patient-derived mitochondrial DNAWe describe the first pathogenic mutation in the mitochondrial ATP8 gene, resulting in an improper assembly and reduced activity of the complex V holoenzyme.

17.
Am J Hum Genet ; 79(5): 869-77, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17033963

ABSTRACT

The 13 polypeptides encoded in mitochondrial DNA (mtDNA) are synthesized in the mitochondrial matrix on a dedicated protein-translation apparatus that resembles that found in prokaryotes. Here, we have investigated the genetic basis for a mitochondrial protein-synthesis defect associated with a combined oxidative phosphorylation enzyme deficiency in two patients, one of whom presented with encephalomyopathy and the other with hypertrophic cardiomyopathy. Sequencing of candidate genes revealed the same homozygous mutation (C997T) in both patients in TSFM, a gene coding for the mitochondrial translation elongation factor EFTs. EFTs functions as a guanine nucleotide exchange factor for EFTu, another translation elongation factor that brings aminoacylated transfer RNAs to the ribosomal A site as a ternary complex with guanosine triphosphate. The mutation predicts an Arg333Trp substitution at an evolutionarily conserved site in a subdomain of EFTs that interacts with EFTu. Molecular modeling showed that the substitution disrupts local subdomain structure and the dimerization interface. The steady-state levels of EFTs and EFTu in patient fibroblasts were reduced by 75% and 60%, respectively, and the amounts of assembled complexes I, IV, and V were reduced by 35%-91% compared with the amounts in controls. These phenotypes and the translation defect were rescued by retroviral expression of either EFTs or EFTu. These data clearly establish mutant EFTs as the cause of disease in these patients. The fact that the same mutation is associated with distinct clinical phenotypes suggests the presence of genetic modifiers of the mitochondrial translation apparatus.


Subject(s)
Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mutation , Peptide Elongation Factors/genetics , Peptide Elongation Factors/metabolism , Amino Acid Sequence , Base Sequence , Cells, Cultured , DNA Primers/genetics , DNA, Mitochondrial/genetics , Electron Transport Chain Complex Proteins/metabolism , Fibroblasts/metabolism , Humans , Mitochondrial Proteins/chemistry , Models, Molecular , Molecular Sequence Data , Multiprotein Complexes , Muscle, Skeletal/metabolism , Oxidative Phosphorylation , Peptide Elongation Factor Tu/chemistry , Peptide Elongation Factor Tu/genetics , Peptide Elongation Factor Tu/metabolism , Peptide Elongation Factors/chemistry , Phenotype , Sequence Homology, Amino Acid
18.
Am J Transplant ; 5(2): 383-93, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15643999

ABSTRACT

Chronic allograft nephropathy (CAN) of renal allografts is still the most important cause of graft loss. A subset of these patients have transplant glomerulopathy (TGP), characterized by glomerular basement membrane (GBM) duplications, but of unknown etiology. Recently, a role for the immune system in the pathogenesis of TGP has been suggested. In 11 of 16 patients with TGP and in 3 of 16 controls with CAN in the absence of TGP we demonstrate circulating antibodies reactive with GBM isolates. The presence of anti-GBM antibodies was associated with the number of rejection episodes prior to diagnosis of TGP. Sera from the TGP patients also reacted with highly purified GBM heparan sulphate proteoglycans (HSPG). Indirect immunofluorescence with patient IgG showed a GBM-like staining pattern and colocalization with the HSPGs perlecan and especially agrin. Using patient IgG, we affinity purified the antigen and identified it as agrin. Reactivity with agrin was found in 7 of 16 (44%) of patients with TGP and in 7 of 11 (64%) patients with anti-GBM reactivity. In conclusion, we have identified a humoral response against the GBM-HSPG agrin in patients with TGP, which may play a role in the pathogenesis of TGP.


Subject(s)
Agrin/immunology , Graft Rejection/immunology , Kidney Glomerulus/immunology , Kidney Transplantation , Antibodies/immunology , Basement Membrane/immunology , Humans
19.
Hum Mol Genet ; 13(20): 2461-72, 2004 Oct 15.
Article in English | MEDLINE | ID: mdl-15317750

ABSTRACT

With 46 subunits, human mitochondrial complex I is the largest enzyme of the oxidative phosphorylation system. We have studied the assembly of complex I in cultured human cells. This will provide essential information about the nature of complex I deficiencies and will enhance our understanding of mitochondrial disease mechanisms. We have found that 143B206 rho zero cells, not containing mitochondrial DNA, are still able to form complex I subcomplexes. To further address the nature of these subcomplexes, we depleted 143B osteosarcoma cells of complex I by inhibiting mitochondrial protein translation with doxycycline. After removing this drug, complex I formation resumes and assembly intermediates were observed by two-dimensional blue native electrophoresis. Analysis of the observed subcomplexes indicates that assembly of human complex I is a semi-sequential process in which different preassembled subcomplexes are joined to form a fully assembled complex. The membrane part of the complex is formed in distinct steps. The B17 subunit is part of a subcomplex to which ND1, ND6 and PSST are subsequently added. This is bound to a hydrophilic subcomplex containing the 30 and 49 kDa subunits, to which a subcomplex including the 39 kDa subunit is incorporated, and later on the 18 and 24 kDa subunits. At a later stage more subunits, including the 15 kDa, are added and holo-complex I is formed. Our results suggest that human complex I assembly resembles that of Neurospora crassa, in which a membrane arm is formed and assembled to a preformed peripheral arm, and support ideas about modular evolution.


Subject(s)
Electron Transport Complex I/metabolism , Mitochondrial Diseases/enzymology , DNA, Mitochondrial/genetics , Doxycycline/pharmacology , Electron Transport Complex I/deficiency , Electron Transport Complex I/genetics , Evolution, Molecular , Humans , Intracellular Membranes/metabolism , Mitochondria/metabolism , Mitochondrial Diseases/genetics , Models, Biological , Osteosarcoma , Protein Transport/drug effects , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...