Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 139
Filtrar
3.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 40(2): 143-147, 2023 Feb 10.
Artigo em Chinês | MEDLINE | ID: mdl-36709930

RESUMO

OBJECTIVE: To carry out preimplantation genetic testing for monogenic/single gene disorders (PGT-M) for a Chinese family affected with Molybdenum co-factor deficiency due to pathogenic variant of MOCS2 gene. METHODS: A family with molybdenum co-factor deficiency who attended to the Maternal and Child Health Care Hospital of Guangxi Zhuang Autonomous Region in April 2020 was selected as the research subject. Trophoblast cells were biopsied from blastocysts fertilized by intracytoplasmic sperm injection. Embryos carrying the MOCS2 gene variant and chromosome copy number variation (CNV) of more than 4 Mb were detected by single-cell whole genome amplification, high-throughput sequencing and single nucleotide polymorphism typing. Embryos without or carrying the heterozygous variant and without abnormal chromosome CNV were transplanted. During mid-pregnancy, amniotic fluid sample was collected for prenatal diagnosis to verify the results of PGT-M. RESULTS: Eleven oocytes were obtained, among which three blastocysts were formed through culturing. Results of genetic testing suggested that one embryo was heterozygous for the maternally derived MOCS2 gene variant and without chromosomal CNV. Following embryo transfer, intrauterine singleton pregnancy was attained. Prenatal diagnosis by amniocentesis at 18 weeks of gestation revealed that the MOCS2 gene variant and chromosomal analysis results were both consistent with that of PGT-M, and a healthy male infant was born at 37+5 weeks of gestation. CONCLUSION: PGT-M has helped the couple carrying the MOCS2 gene variant to have a healthy offspring, and may become an important method for couples carrying other pathogenic genetic variants.


Assuntos
Erros Inatos do Metabolismo dos Metais , Diagnóstico Pré-Implantação , Feminino , Humanos , Gravidez , Aneuploidia , China , Variações do Número de Cópias de DNA , Testes Genéticos/métodos , Diagnóstico Pré-Implantação/métodos , Erros Inatos do Metabolismo dos Metais/genética
4.
Artigo em Chinês | WPRIM (Pacífico Ocidental) | ID: wpr-970894

RESUMO

OBJECTIVE@#To carry out preimplantation genetic testing for monogenic/single gene disorders (PGT-M) for a Chinese family affected with Molybdenum co-factor deficiency due to pathogenic variant of MOCS2 gene.@*METHODS@#A family with molybdenum co-factor deficiency who attended to the Maternal and Child Health Care Hospital of Guangxi Zhuang Autonomous Region in April 2020 was selected as the research subject. Trophoblast cells were biopsied from blastocysts fertilized by intracytoplasmic sperm injection. Embryos carrying the MOCS2 gene variant and chromosome copy number variation (CNV) of more than 4 Mb were detected by single-cell whole genome amplification, high-throughput sequencing and single nucleotide polymorphism typing. Embryos without or carrying the heterozygous variant and without abnormal chromosome CNV were transplanted. During mid-pregnancy, amniotic fluid sample was collected for prenatal diagnosis to verify the results of PGT-M.@*RESULTS@#Eleven oocytes were obtained, among which three blastocysts were formed through culturing. Results of genetic testing suggested that one embryo was heterozygous for the maternally derived MOCS2 gene variant and without chromosomal CNV. Following embryo transfer, intrauterine singleton pregnancy was attained. Prenatal diagnosis by amniocentesis at 18 weeks of gestation revealed that the MOCS2 gene variant and chromosomal analysis results were both consistent with that of PGT-M, and a healthy male infant was born at 37+5 weeks of gestation.@*CONCLUSION@#PGT-M has helped the couple carrying the MOCS2 gene variant to have a healthy offspring, and may become an important method for couples carrying other pathogenic genetic variants.


Assuntos
Feminino , Humanos , Gravidez , Aneuploidia , China , Variações do Número de Cópias de DNA , Testes Genéticos/métodos , Diagnóstico Pré-Implantação/métodos , Erros Inatos do Metabolismo dos Metais/genética
5.
Neuropediatrics ; 53(3): 200-203, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-34674206

RESUMO

BACKGROUND: To describe the genotype-phenotype dissociation in two Taiwanese patients with molybdenum cofactor deficiency (MoCoD) caused by MOCS2 gene mutations. PATIENT DESCRIPTION: Patient 1 exhibited early-onset neurological symptoms soon after birth, followed by subsequent myoclonic seizures and movement disorder. The brain magnetic resonance imaging (MRI) showed diffuse brain injury with cystic encephalomalacia along with bilateral globus pallidi involvement, hypoplasia of corpus callosum, and cerebellar atrophy. Patient 2 had a mild phenotype with prominent movement disorder after intercurrent illness, and the brain MRI showed selective injury of the bilateral globus pallidi and the cerebellum. Both patients had markedly low levels of plasma uric acid and harbored the same MOCS2 homozygous c.16C > T mutation. Patient 1 showed chronic regression of developmental milestones and died of respiratory failure at the age of 8 years, whereas patient 2 demonstrated improvement in motor function. CONCLUSION: Genotype-phenotype dissociation could be noted in patients with MoCoD due to MOCS2 mutation. Patients with neonatal seizures, developmental delay, movement disorder, and motor regression after an illness, as well as focal or bilateral involvement of the globus pallidi on the neuroimages, should undergo biochemical testing of plasma uric acid. A pronounced plasma uric acid level is a good indicator of MoCoD. Early diagnosis can allow early provision of adequate genetic counseling.


Assuntos
Erros Inatos do Metabolismo dos Metais , Transtornos dos Movimentos , Ácido Úrico , Criança , Homozigoto , Humanos , Imageamento por Ressonância Magnética , Erros Inatos do Metabolismo dos Metais/genética , Transtornos dos Movimentos/complicações , Mutação , Fenótipo , Convulsões/genética , Sulfurtransferases/genética
6.
Zhonghua Er Ke Za Zhi ; 59(2): 119-124, 2021 Feb 02.
Artigo em Chinês | MEDLINE | ID: mdl-33548958

RESUMO

Objective: To explore the phenotypes and genotypes of molybdenum cofactor deficiency type B (MoCD-B) manifested as Leigh-like syndrome. Methods: The clinical data, laboratory tests, neuroimaging and gene results of one patient diagnosed as MoCD-B at Beijing Children's Hospital and Hebei Children's Hospital in December 2018 were collected. Related literature was searched and reviewed at Wanfang Data Knowledge Service Platform, China National Knowledge Infrastructure and PubMed (up to September 2020) by using terms "MOCS2" "molybdenum cofactor deficiency" "Leigh-like syndrome,MOCS2" "molybdenum cofactor deficiency, Leigh-like syndrome". The phenotypes and genotypes of MoCD-B were summarized. Results: A 7 months and 14 days old boy with the chief complaint of "cough for 6 days, abnormal posture for 4 days and fever for 2 days" was admitted to Hebei Children' Hospital on December 2018. His abnormal posture presented as opisthotonos accompanied with dysphagia, without seizures. His previous psychomotor development was described as normal. He was born at term after an uneventful pregnancy to non-consanguineous parents. Blood test showed a slightly increased lactic acid and a significantly decreased uric acid. Urine metabolism test showed an obviously increased xanthine and hypoxanthine. Brain magnetic resonance imaging showed hyperintense signal on T2 weighted image and fluid attenuated inversion recovery in bilateral globus pallidus and pedunculus cerebri. The patient was diagnosed with Leigh-like syndrome. No obvious improvement was achieved after cocktail therapy and symptomatic treatment. The whole exome sequencing showed that the patient carried a homozygous variant of MOCS2 gene, c.19G>T(p.Val7Phe), which was a previously reported pathogenic site in the literature and could cause MoCD-B. His parents carried a heterozygous variant respectively. A total of 41 MoCD-B cases with MOCS2 gene variants were collected through literature review and our study, among which 30 cases had full medical records. The onset ages of 23 (77%) cases were in neonate, manifesting with severe encephalopathy, including neonatal-onset intractable seizures, developmental delay, laboratory abnormalities included very low levels of serum and urinary uric acid, increased urinary levels of xanthine and hypoxanthine. Cranial imaging showed cerebral atrophy, cystic encephalomalacia, etc. The onset ages of 7 patients varied from 5 months to 23 years. Four cases had normal psychomotor development before disease onset. Neurological disorders appeared acutely or exacerbated after external triggers and all of them had basal ganglia involvement. Among the 30 cases, 3 cases had a relatively milder phenotype with the ability of brief communication and walking without or with support. Conclusions: Molybdenum cofactor deficiency is a rare disease. Most cases had severe phenotypes and poor outcomes, but some cases may have mild phenotype. MoCD-B caused by MOCS2 gene variants may manifest as Leigh-like syndrome with a normal psychomotor development before the trigger of infection strike. Hypouricemia, xanthinuria and hypoxanthinuria can be indicators of the disease. The presence of MOCS2 gene variants would confirm a final diagnosis.


Assuntos
Erros Inatos do Metabolismo dos Metais , Criança , Pré-Escolar , China , Homozigoto , Humanos , Lactente , Recém-Nascido , Masculino , Erros Inatos do Metabolismo dos Metais/diagnóstico , Erros Inatos do Metabolismo dos Metais/genética , Fenótipo
7.
J Dermatol ; 48(4): 519-528, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33458872

RESUMO

PSTPIP1-associated myeloid-related proteinaemia inflammatory (PAMI) syndrome has been described as a rare and distinct clinical phenotype of PSTPIP1-associated inflammatory diseases. We report PSTPIP1 mutation in both father and son who have leukopenia and acne-like lesions. Through whole-exome sequencing on blood DNA, it is found a heterozygous mutation of PSTPIP1 gene c.748G>A on the father and son. The diagnosis of PAMI is made based on DNA sequencing results and clinical characteristics of typical lesions, leukopenia, and the markedly increased serum S100A8/A9 (calprotectin).


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas do Citoesqueleto/genética , Pai , Humanos , Inflamação/genética , Masculino , Erros Inatos do Metabolismo dos Metais/genética , Mutação , Mutação de Sentido Incorreto , Síndrome
9.
Hematology Am Soc Hematol Educ Program ; 2020(1): 465-470, 2020 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-33275715

RESUMO

Inherited microcytic anemias can be broadly classified into 3 subgroups: (1) defects in globin chains (hemoglobinopathies or thalassemias), (2) defects in heme synthesis, and (3) defects in iron availability or iron acquisition by the erythroid precursors. These conditions are characterized by a decreased availability of hemoglobin (Hb) components (globins, iron, and heme) that in turn causes a reduced Hb content in red cell precursors with subsequent delayed erythroid differentiation. Iron metabolism alterations remain central to the diagnosis of microcytic anemia, and, in general, the iron status has to be evaluated in cases of microcytosis. Besides the very common microcytic anemia due to acquired iron deficiency, a range of hereditary abnormalities that result in actual or functional iron deficiency are now being recognized. Atransferrinemia, DMT1 deficiency, ferroportin disease, and iron-refractory iron deficiency anemia are hereditary disorders due to iron metabolism abnormalities, some of which are associated with iron overload. Because causes of microcytosis other than iron deficiency should be considered, it is important to evaluate several other red blood cell and iron parameters in patients with a reduced mean corpuscular volume (MCV), including mean corpuscular hemoglobin, red blood cell distribution width, reticulocyte hemoglobin content, serum iron and serum ferritin levels, total iron-binding capacity, transferrin saturation, hemoglobin electrophoresis, and sometimes reticulocyte count. From the epidemiological perspective, hemoglobinopathies/thalassemias are the most common forms of hereditary microcytic anemia, ranging from inconsequential changes in MCV to severe anemia syndromes.


Assuntos
Anemia , Ferro/metabolismo , Erros Inatos do Metabolismo dos Metais , Anemia/classificação , Anemia/genética , Anemia/metabolismo , Anemia/patologia , Humanos , Erros Inatos do Metabolismo dos Metais/classificação , Erros Inatos do Metabolismo dos Metais/genética , Erros Inatos do Metabolismo dos Metais/metabolismo , Erros Inatos do Metabolismo dos Metais/patologia
10.
Eur J Med Genet ; 63(5): 103874, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32028041

RESUMO

Congenital atransferrinemia is an extremely rare autosomal recessive disorder resulting in the complete absence or extremely reduced amount of transferrin. In this study, we describe the first case of congenital atransferrinemia in Tunisia and the 18th patient in the reported data. The patient was referred to our hospital to explore a severe hypochromic and microcytic anemia. The laboratory evaluation including hematological and biochemical examination was performed in the proband and her parents. All exons of the transferrin gene were PCR amplified. The products were screened for mutations by direct sequencing. Based on laboratory and clinical findings, diagnosis of congenital atransferrinemia was confirmed. DNA sequencing revealed the presence of a novel homozygous deletion (c.293-63del) in the intron 13. This mutation is predicted to generate a higher score cryptic branch point leading to the production of an altered mRNA molecule. The second previously reported missense mutation p.Arg609Trp. Crystallographic structure analyzes demonstrate that the mutation would probably lead to significant conformational change not allowing the expression of transferrin protein. Current molecular characterization of this novel transferrin abnormality puts to the proof the variability in onset, first blood transfusion, and phenotypic expression in atransferrinemic patients.


Assuntos
Erros Inatos do Metabolismo dos Metais/genética , Mutação , Sítios de Splice de RNA , Transferrina/deficiência , Transferrina/genética , Feminino , Homozigoto , Humanos , Lactente , Erros Inatos do Metabolismo dos Metais/patologia , Domínios Proteicos , Transferrina/química , Transferrina/metabolismo
11.
Pediatr Neurol ; 99: 55-59, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31201073

RESUMO

BACKGROUND: We explored the clinical and molecular characteristics of molybdenum cofactor deficiency due to MOCS2 muations. METHODS: We summarize the genetic and clinical findings of previously reported patients with a MOCS2 mutation. We also present a new patient with novel neuroradiological findings associated with molybdenum cofactor deficiency due to a novel homozygous variant in the 5' untranslated region of the MOCS2 gene. RESULTS: The study population comprised 35 patients with a MOCS2 gene mutation. All reported children had delayed motor milestones. The major initial symptom was seizures in neonatal period. Facial dysmorphism was present in 61% of the patients. Only one patient had ectopia lentis. Agenesis of the corpus callosum and an associated interhemispheric cyst in our case are novel neuroradiological findings. CONCLUSIONS: The occurrence of neonatal seizures and feeding difficulties can be the first clinical signs of molybdenum cofactor deficiency. Although there is no effective therapy for this condition, early diagnosis and genetic analysis of these lethal disorders facilitate adequate genetic counseling.


Assuntos
Erros Inatos do Metabolismo dos Metais/genética , Sulfurtransferases/deficiência , Regiões 5' não Traduzidas/genética , Agenesia do Corpo Caloso/diagnóstico por imagem , Agenesia do Corpo Caloso/genética , Cisterna Magna/diagnóstico por imagem , Cisterna Magna/patologia , Bases de Dados Factuais , Encefalomalacia/diagnóstico por imagem , Encefalomalacia/genética , Face/anormalidades , Transtornos de Alimentação na Infância/genética , Feminino , Heterogeneidade Genética , Homozigoto , Humanos , Lactente , Recém-Nascido , Imageamento por Ressonância Magnética , Masculino , Transtornos dos Movimentos/congênito , Transtornos dos Movimentos/genética , Neuroimagem , Fenótipo , Convulsões/congênito , Sulfurtransferases/genética , Substância Branca/diagnóstico por imagem , Substância Branca/patologia
12.
Mol Genet Genomic Med ; 7(6): e657, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30900395

RESUMO

BACKGROUND: Molybdenum cofactor deficiency (MoCD) is a rare autosomal-recessive disorder that results in the combined deficiency of molybdenum-dependent enzymes. Four different genes are involved in Molybdenum cofactor biosynthesis: MOCS1, MOCS2, MOCS3, and GEPH. The classical form manifests in the neonatal period with severe encephalopathy, including intractable seizures, MRI changes that resemble hypoxic-ischemic injury, microcephaly, and early death. To date, an atypical phenotype with late-onset has been reported in the literature in 13 patients. METHODS: We describe a late-onset and a relatively mild phenotype in a patient with MOCS2 homozygous mutation. RESULTS: Pyramidal and extrapyramidal signs are recognized in those patients, often exacerbated by intercurrent illness. Expressive language is usually compromised. Neurological deterioration is possible even in adulthood, probably due to accumulation of sulfite with time. CONCLUSION: Sulfite inhibition of mitochondrial metabolism could be responsible for the ischemic lesions described in patients with MoCD or alternatively could predispose the brain to suffer an ischemic damage through the action of other insults, for instance intercurrent illness. It is possible that sulfite accumulation together with other external triggers, can lead to neurological deterioration even in adulthood. The role of other factors involved in clinical expression should be investigated to establish the reason for phenotypic variability in patients with the same mutation.


Assuntos
Erros Inatos do Metabolismo dos Metais/genética , Erros Inatos do Metabolismo dos Metais/fisiopatologia , Sulfurtransferases/genética , Criança , Coenzimas/genética , Coenzimas/metabolismo , Feminino , Homozigoto , Humanos , Erros Inatos do Metabolismo dos Metais/metabolismo , Metaloproteínas/genética , Metaloproteínas/metabolismo , Cofatores de Molibdênio , Mutação , Fenótipo , Pteridinas/metabolismo , Sulfurtransferases/metabolismo
13.
Hum Genet ; 138(4): 355-361, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30810871

RESUMO

Molybdenum cofactor deficiency is an autosomal, recessively inherited metabolic disorder, which, in the absence of an effective therapy, leads to early childhood death due to neurological deterioration. In type A of the disease, cyclic pyranopterin monophosphate (cPMP) is missing, the first intermediate in the biosynthesis of the cofactor, and a biochemical substitution therapy using cPMP has been developed. A comparable approach for type B of the disease with a defect in the second step of the synthesis, formation of molybdopterin, so far has been hampered by the extreme instability of the corresponding metabolites. To explore avenues for a successful and safe gene therapy, knock-in mouse models were created carrying the mutations c.88C>T (p.Q30X) and c.726_727delAA, which are also found in human patients. Recombinant adeno-associated viruses (rAAVs) were constructed and used for postnatal intrahepatic injections of MoCo-deficient mice in a proof-of-concept approach. Singular administration of an appropriate virus dose in 60 animals prevented the otherwise devastating phenotype to a variable extent. While untreated mice did not survive for more than 2 weeks, some of the treated mice grew up to adulthood in both sexes.


Assuntos
Dependovirus , Terapia Genética/métodos , Erros Inatos do Metabolismo dos Metais/genética , Erros Inatos do Metabolismo dos Metais/terapia , Sulfurtransferases/genética , Animais , Dependovirus/genética , Modelos Animais de Doenças , Terapia de Reposição de Enzimas/métodos , Estudos de Viabilidade , Técnicas de Introdução de Genes , Humanos , Injeções , Erros Inatos do Metabolismo dos Metais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Sulfurtransferases/administração & dosagem
14.
Neuropediatrics ; 50(2): 126-129, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30695801

RESUMO

Molybdenum cofactor deficiency (MoCD) is a rare autosomal recessive metabolic disease with severe neurological symptoms. Most disease-causing mutations are found in the MOCS1 gene, corresponding to MoCD type A (MoCD-A). There have been few reports describing the long-term detailed neurological features with MoCD-A because most patients do not survive childhood. We describe the clinical, radiologic, biochemical, and genetic data of two patients (female siblings aged 26 and 22 years) with MoCD-A. Both patients presented with feeding difficulties, neurological deterioration, and persistent generalized muscle contraction which can be easily confused with status dystonicus. Biochemical tests revealed low serum uric acid, elevated urinary sulfocysteine, and xanthine. Brain magnetic resonance imaging (MRI) revealed distinctive abnormalities in the bilateral caudate nucleus, putamen, globus pallidus, and cerebral white matter adjacent to the cortex. The thalamus was relatively unaffected. Genetic testing identified a novel homozygous variant in the MOCS1 gene (c.949C > T p.Arg317Cys). Biochemical results supported the hypothesis that this genetic variant is a pathological mutation. When there are symptoms of persistent generalized muscle contraction and characteristic MRI findings, MoCD should be considered as a differential diagnosis.


Assuntos
Erros Inatos do Metabolismo dos Metais/diagnóstico por imagem , Erros Inatos do Metabolismo dos Metais/genética , Contração Muscular/genética , Irmãos , Adulto , Feminino , Humanos , Erros Inatos do Metabolismo dos Metais/complicações , Adulto Jovem
15.
Mol Genet Metab ; 124(2): 161-167, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29685658

RESUMO

Congenital disorders of manganese metabolism are rare occurrences in children, and medical management of these disorders is complex and challenging. Homozygous exonic mutations in the manganese transporter SLC39A14 have recently been associated with a pediatric-onset neurodegenerative disorder characterized by brain manganese accumulation and clinical signs of manganese neurotoxicity, including parkinsonism-dystonia. We performed whole exome sequencing on DNA samples from two unrelated female children from the United Arab Emirates with progressive movement disorder and brain mineralization, identified a novel homozygous intronic mutation in SLC39A14 in both children, and demonstrated that the mutation leads to aberrant splicing. Both children had consistently elevated serum manganese levels and were diagnosed with SLC39A14-associated manganism. Over a four-year period, we utilized a multidisciplinary management approach for Patient 1 combining decreased manganese dietary intake and chelation with symptomatic management of dystonia. Our treatment strategy appeared to slow disease progression, but did not lead to a cure or reversal of already established deficits. Clinicians should consider testing for noncoding mutations in the diagnosis of congenital disorders of manganese metabolism and utilizing multidisciplinary approaches in the management of these disorders.


Assuntos
Proteínas de Transporte de Cátions/genética , Distúrbios Distônicos/genética , Manganês/metabolismo , Erros Inatos do Metabolismo dos Metais/genética , Mutação , Transtornos Parkinsonianos/genética , Quelantes/uso terapêutico , Criança , Pré-Escolar , Distúrbios Distônicos/tratamento farmacológico , Distúrbios Distônicos/patologia , Feminino , Humanos , Masculino , Erros Inatos do Metabolismo dos Metais/tratamento farmacológico , Erros Inatos do Metabolismo dos Metais/patologia , Transtornos Parkinsonianos/tratamento farmacológico , Transtornos Parkinsonianos/patologia , Linhagem
17.
J Vet Intern Med ; 32(2): 853-859, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29424482

RESUMO

BACKGROUND: Zinc deficiency-like (ZDL) syndrome is an inherited defect of Fleckvieh calves, with striking similarity to bovine hereditary zinc deficiency (BHZD). However, the causative mutation in a phospholipase D4 encoding gene (PLD4) shows no connection to zinc metabolism. OBJECTIVES: To describe clinical signs, laboratory variables, and pathological findings of ZDL syndrome and their utility to differentiate ZDL from BHZD and infectious diseases with similar phenotype. ANIMALS: Nine hospitalized calves with crusting dermatitis and confirmed mutation in PLD4 and medical records from 25 calves with crusting dermatitis or suspected zinc deficiency. METHODS: Prospective and retrospective case series. RESULTS: The 9 calves (age: 5-53 weeks) displayed a moderate to severe crusting dermatitis mainly on the head, ventrum, and joints. Respiratory and digestive tract inflammations were frequently observed. Zinc supplementation did not lead to remission of clinical signs in 4 calves. Laboratory variables revealed slight anemia in 8 calves, hypoalbuminemia in 6 calves, but reduced serum zinc concentrations in only 3 calves. Mucosal erosions/ulcerations were present in 7 calves and thymus atrophy or reduced thymic weights in 8 calves. Histologically, skin lesions were indistinguishable from BHZD. Retrospective analysis of medical records revealed the presence of this phenotype since 1988 and pedigree analysis revealed a common ancestor of several affected calves. CONCLUSIONS AND CLINICAL IMPORTANCE: ZDL syndrome should be suspected in Fleckvieh calves with crusting dermatitis together with diarrhea or respiratory tract inflammations without response to oral zinc supplementation. Definite diagnosis requires molecular genetic confirmation of the PLD4 mutation.


Assuntos
Doenças dos Bovinos/patologia , Zinco/sangue , Animais , Bovinos , Doenças dos Bovinos/diagnóstico , Doenças dos Bovinos/genética , Dermatite/diagnóstico , Dermatite/genética , Dermatite/veterinária , Feminino , Masculino , Erros Inatos do Metabolismo dos Metais/diagnóstico , Erros Inatos do Metabolismo dos Metais/genética , Estudos Prospectivos , Estudos Retrospectivos , Síndrome , Zinco/uso terapêutico
18.
J Inherit Metab Dis ; 41(2): 187-196, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29368224

RESUMO

Molybdenum cofactor deficiency is an autosomal recessive inborn error of metabolism, which results from mutations in genes involved in Moco biosynthesis. Moco serves as a cofactor of several enzymes, including sulfite oxidase. MoCD is clinically characterized by intractable seizures and severe, rapidly progressing neurodegeneration leading to death in early childhood in the majority of known cases. Here we report a patient with an unusual late disease onset and mild phenotype, characterized by a lack of seizures, normal early development, a decline triggered by febrile illness and a subsequent dystonic movement disorder. Genetic analysis revealed a homozygous c.1338delG MOCS1 mutation causing a frameshift (p.S442fs) with a premature termination of the MOCS1AB translation product at position 477 lacking the entire MOCS1B domain. Surprisingly, urine analysis detected trace amounts (1% of control) of the Moco degradation product urothione, suggesting a residual Moco synthesis in the patient, which was consistent with the mild clinical presentation. Therefore, we performed bioinformatic analysis of the patient's mutated MOCS1 transcript and found a potential Kozak-sequence downstream of the mutation site providing the possibility of an independent expression of a MOCS1B protein. Following the expression of the patient's MOCS1 cDNA in HEK293 cells we detected two proteins: a truncated MOCS1AB protein and a 22.4 kDa protein representing MOCS1B. Functional studies of both proteins confirmed activity of MOCS1B, but not of the truncated MOCS1AB. This finding demonstrates an unusual mechanism of translation re-initiation in the MOCS1 transcript, which results in trace amounts of functional MOCS1B protein being sufficient to partially protect the patient from the most severe symptoms of MoCD.


Assuntos
Coenzimas/metabolismo , Erros Inatos do Metabolismo dos Metais/metabolismo , Metaloproteínas/metabolismo , Proteínas Nucleares/metabolismo , Fragmentos de Peptídeos/metabolismo , Pteridinas/metabolismo , Idade de Início , Carbono-Carbono Liases , Criança , Pré-Escolar , Dieta com Restrição de Proteínas , Mutação da Fase de Leitura , Predisposição Genética para Doença , Células HEK293 , Humanos , Imageamento por Ressonância Magnética , Masculino , Erros Inatos do Metabolismo dos Metais/diagnóstico , Erros Inatos do Metabolismo dos Metais/dietoterapia , Erros Inatos do Metabolismo dos Metais/genética , Cofatores de Molibdênio , Proteínas Nucleares/genética , Fragmentos de Peptídeos/genética , Fenótipo
19.
Clin Exp Immunol ; 191(2): 240-251, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28984903

RESUMO

A child, 2 years with the 'hypercalprotectinaemia with hyperzincaemia' clinical syndrome, presented with atypical symptoms and signs, notably persistent fever of approximately 38°C, thrombocythaemia of > 700 × 109 /l and a predominance of persistent intestinal symptoms. In an effort to find a cure by identifying the dysregulated pathways we analysed whole-genome mRNA expression by the Affymetrix HG U133 Plus 2·0 array in blood on three occasions 3-5 months apart. Major up-regulation was demonstrated for the Janus kinase/signal transducer and activators of transcription (JAK/STAT) pathway including, in particular, CD177, S100A8, S100A9 and S100A12, accounting for the thrombocytosis; a large number of interleukins, their receptors and activators, accounting for the febrile apathic state; and the high mobility group box 1 (HMBG1) gene, possibly accounting for part of the intestinal symptoms. These results show that gene expression array technology may assist the clinician in the diagnostic work-up of individual patients with suspected syndromal states of unknown origin, and the expression data can guide the selection of optimal treatment directed at the identified target pathways.


Assuntos
Proteínas Sanguíneas/genética , Complexo Antígeno L1 Leucocitário/metabolismo , Erros Inatos do Metabolismo dos Metais/diagnóstico , Pré-Escolar , Tomada de Decisão Clínica , Feminino , Febre , Gastroenteropatias , Estudo de Associação Genômica Ampla , Humanos , Complexo Antígeno L1 Leucocitário/genética , Erros Inatos do Metabolismo dos Metais/genética , Análise em Microsséries , Patologia Molecular , Transdução de Sinais , Trombocitose , Sequenciamento Completo do Genoma
20.
J Inherit Metab Dis ; 40(6): 801-811, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28900816

RESUMO

INTRODUCTION: Molybdenum cofactor deficiency (MoCD) is an ultra-orphan, life-threatening disease. Substrate substitution therapy has successfully been performed in single cases of MoCD type A and clinical trials are underway for drug registration. We present an innovative approach for classification of genotype severity to test the hypothesis that milder sequence variants in MoCD result in a less severe disease phenotype quantitated by patient survival. METHODS: All available worldwide published cases with clinical and genetic data were included (n = 40). We stratified the already published disease causing sequence variants as mild or severe with the use of in silico prediction programs, where possible and assessed the possible impact of the variants on the expression of the gene or function of the expressed protein. In a compound heterozygous situation the mildest sequence variant determined the genotype. Subsequently, clinical manifestations and outcomes of both groups were compared. RESULTS: Patients with a severe genotype showed a median survival of 15 months and had a lower probability of survival compared to patients with mild genotypes who were all alive at last reported follow-up (p = 0.0203, Log-rank test). DISCUSSION: The severity of the genotype assessed by in silico prediction and further classification explained survival in molybdenum cofactor deficiency and may therefore be considered a confounder for the outcome of therapeutic clinical trials requiring adjustment in the clinical trial design or analysis. These results should further be investigated by future in vitro or in vivo functional studies. Caution should be taken with this approach for the classification of variants in molecular genetic diagnostics or genetic counseling.


Assuntos
Erros Inatos do Metabolismo dos Metais/genética , Adolescente , Adulto , Criança , Pré-Escolar , Feminino , Variação Genética/genética , Genótipo , Humanos , Lactente , Recém-Nascido , Masculino , Fenótipo , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...