Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
N Engl J Med ; 389(7): 620-631, 2023 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-37585628

RESUMO

BACKGROUND: Patients with the Crigler-Najjar syndrome lack the enzyme uridine diphosphoglucuronate glucuronosyltransferase 1A1 (UGT1A1), the absence of which leads to severe unconjugated hyperbilirubinemia that can cause irreversible neurologic injury and death. Prolonged, daily phototherapy partially controls the jaundice, but the only definitive cure is liver transplantation. METHODS: We report the results of the dose-escalation portion of a phase 1-2 study evaluating the safety and efficacy of a single intravenous infusion of an adeno-associated virus serotype 8 vector encoding UGT1A1 in patients with the Crigler-Najjar syndrome that was being treated with phototherapy. Five patients received a single infusion of the gene construct (GNT0003): two received 2×1012 vector genomes (vg) per kilogram of body weight, and three received 5×1012 vg per kilogram. The primary end points were measures of safety and efficacy; efficacy was defined as a serum bilirubin level of 300 µmol per liter or lower measured at 17 weeks, 1 week after discontinuation of phototherapy. RESULTS: No serious adverse events were reported. The most common adverse events were headache and alterations in liver-enzyme levels. Alanine aminotransferase increased to levels above the upper limit of the normal range in four patients, a finding potentially related to an immune response against the infused vector; these patients were treated with a course of glucocorticoids. By week 16, serum bilirubin levels in patients who received the lower dose of GNT0003 exceeded 300 µmol per liter. The patients who received the higher dose had bilirubin levels below 300 µmol per liter in the absence of phototherapy at the end of follow-up (mean [±SD] baseline bilirubin level, 351±56 µmol per liter; mean level at the final follow-up visit [week 78 in two patients and week 80 in the other], 149±33 µmol per liter). CONCLUSIONS: No serious adverse events were reported in patients treated with the gene-therapy vector GNT0003 in this small study. Patients who received the higher dose had a decrease in bilirubin levels and were not receiving phototherapy at least 78 weeks after vector administration. (Funded by Genethon and others; ClinicalTrials.gov number, NCT03466463.).


Assuntos
Síndrome de Crigler-Najjar , Terapia Genética , Glucuronosiltransferase , Humanos , Administração Intravenosa , Bilirrubina/sangue , Síndrome de Crigler-Najjar/sangue , Síndrome de Crigler-Najjar/complicações , Síndrome de Crigler-Najjar/genética , Síndrome de Crigler-Najjar/terapia , Dependovirus , Terapia Genética/efeitos adversos , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Glucuronosiltransferase/administração & dosagem , Glucuronosiltransferase/genética , Hiperbilirrubinemia/sangue , Hiperbilirrubinemia/etiologia , Hiperbilirrubinemia/genética , Hiperbilirrubinemia/terapia , Transplante de Fígado , Fototerapia
2.
JIMD Rep ; 52(1): 17-22, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32154055

RESUMO

Glycogen storage disease type Ia (GSD Ia) is a rare metabolic disease due to glucose-6-phosphatase deficiency. Chronic kidney disease is a frequent complication that may manifest itself by glomerular lesions and tubular dysfunction from the second decade of life. We report two young GSDIa patients with malignant renal tumor. The first patient was a 25-year-old man. He had chronic metabolic imbalance without kidney involvement. The tumor, a type 2 papillary renal carcinoma, was accidentally discovered during follow-up. The second patient was a 27-year-old woman with chronic metabolic imbalance and chronic kidney involvement. The tumor, a grade 2 papillary carcinoma, was accidentally discovered during follow-up. These two observations are, to date, the first to be reported. We suggest that annual monitoring of kidney imaging in GSDI patients should be systematic to detect renal cancer, from the second decade of life.

3.
Xenotransplantation ; 27(1): e12544, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31342573

RESUMO

Pluripotent stem cells have been investigated as a renewable source of therapeutic hepatic cells, in order to overcome the lack of transplantable donor hepatocytes. Whereas different studies were able to correct hepatic defects in animal models, they focused on the most mature phenotype of hepatocyte-like cells (HLCs) derived from pluripotent stem cells and needed freshly prepared cells, which limits clinical applications of HLCs. Here, we report the production of hepatic stem cells (pHSCs) from human-induced pluripotent stem cells (hiPSCs) in xeno-free, feeder-free, and chemically defined conditions using as extracellular matrix a recombinant laminin instead of Matrigel, an undefined animal-derived matrix. Freshly prepared and frozen pHSCs were transplanted via splenic injection in Gunn rats, the animal model for Crigler-Najjar syndrome. Following cell transplantation and daily immunosuppression treatment, bilirubinemia was significantly decreased (around 30% decrease, P < .05) and remained stable throughout the 6-month study. The transplanted pHSCs underwent maturation in vivo to restore the deficient metabolic hepatic function (bilirubin glucuronidation by UGT1A1). In conclusion, we demonstrate for the first time the differentiation of hiPSCs into pHSCs that (a) are produced using a differentiation protocol compatible with Good Manufacturing Practices, (b) can be frozen, and (c) are sufficient to demonstrate in vivo therapeutic efficacy to significantly lower hyperbilirubinemia in a model of inherited liver disease, despite their immature phenotype. Thus, our approach provides major advances toward future clinical applications and would facilitate cell therapy manufacturing from human pluripotent stem cells.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Síndrome de Crigler-Najjar/terapia , Hepatócitos/fisiologia , Hiperbilirrubinemia/terapia , Células-Tronco Pluripotentes Induzidas/fisiologia , Fígado/fisiologia , Transplante de Células-Tronco/métodos , Animais , Diferenciação Celular , Células Cultivadas , Criopreservação , Modelos Animais de Doenças , Humanos , Fígado/cirurgia , Ratos , Ratos Gunn , Medicina Regenerativa/métodos , Transplante Heterólogo
4.
Acta Neuropathol Commun ; 7(1): 167, 2019 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-31661040

RESUMO

Glycogen storage disorder type III (GSDIII), or debranching enzyme (GDE) deficiency, is a rare metabolic disorder characterized by variable liver, cardiac, and skeletal muscle involvement. GSDIII manifests with liver symptoms in infancy and muscle involvement during early adulthood. Muscle biopsy is mainly performed in patients diagnosed in adulthood, as routine diagnosis relies on blood or liver GDE analysis, followed by AGL gene sequencing. The GSDIII mouse model recapitulate the clinical phenotype in humans, and a nearly full rescue of muscle function was observed in mice treated with the dual AAV vector expressing the GDE transgene.In order to characterize GSDIII muscle morphological spectrum and identify novel disease markers and pathways, we performed a large international multicentric morphological study on 30 muscle biopsies from GSDIII patients. Autophagy flux studies were performed in human muscle biopsies and muscles from GSDIII mice. The human muscle biopsies revealed a typical and constant vacuolar myopathy, characterized by multiple and variably sized vacuoles filled with PAS-positive material. Using electron microscopy, we confirmed the presence of large non-membrane bound sarcoplasmic deposits of normally structured glycogen as well as smaller rounded sac structures lined by a continuous double membrane containing only glycogen, corresponding to autophagosomes. A consistent SQSTM1/p62 decrease and beclin-1 increase in human muscle biopsies suggested an enhanced autophagy. Consistent with this, an increase in the lipidated form of LC3, LC3II was found in patients compared to controls. A decrease in SQSTM1/p62 was also found in the GSDIII mouse model.In conclusion, we characterized the morphological phenotype in GSDIII muscle and demonstrated dysfunctional autophagy in GSDIII human samples.These findings suggest that autophagic modulation combined with gene therapy might be considered as a novel treatment for GSDIII.


Assuntos
Autofagia , Doença de Depósito de Glicogênio Tipo III/patologia , Músculo Esquelético/patologia , Vacúolos/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Biópsia , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Músculo Esquelético/ultraestrutura
5.
Hum Gene Ther ; 30(10): 1297-1305, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31502485

RESUMO

Adeno-associated virus (AAV) vector-mediated gene therapy is currently evaluated as a potential treatment for Crigler-Najjar syndrome (CN) (NCT03466463). Pre-existing immunity to AAV is known to hinder gene transfer efficacy, restricting enrollment of seropositive subjects in ongoing clinical trials. We assessed the prevalence of anti-AAV serotype 8 (AAV8) neutralizing antibodies (NAbs) in subjects affected by CN and investigated the impact of low NAb titers (<1:5) on liver gene transfer efficacy in an in vivo passive immunization model. A total of 49 subjects with a confirmed molecular diagnosis of CN were included in an international multicenter study (NCT02302690). Pre-existing NAbs against AAV8 were detected in 30.6% (15/49) of screened patients and, in the majority of positive cases, cross-reactivity to AAV2 and AAV5 was detected. To investigate the impact of low NAbs on AAV vector-mediated liver transduction efficiency, adult wild-type C57BL/6 mice were passively immunized with pooled human donor-derived immunoglobulins to achieve titers of up to 1:3.16. After immunization, animals were injected with different AAV8 vector preparations. Hepatic vector gene copy number was unaffected by low anti-AAV8 NAb titers when column-purified AAV vector batches containing both full and empty capsids were used. In summary, although pre-existing anti-AAV8 immunity can be found in about a third of subjects affected by CN, low anti-AAV8 NAb titers are less likely to affect liver transduction efficiency when using AAV vector preparations manufactured to contain both full and empty capsids. These findings have implications for the design of liver gene transfer clinical trials and for the definition of inclusion criteria related to seropositivity of potential participants.


Assuntos
Anticorpos Neutralizantes/biossíntese , Anticorpos Antivirais/biossíntese , Síndrome de Crigler-Najjar/terapia , Dependovirus/genética , Terapia Genética/métodos , Glucuronosiltransferase/genética , Adolescente , Adulto , Animais , Bilirrubina/imunologia , Bilirrubina/metabolismo , Capsídeo/imunologia , Capsídeo/metabolismo , Criança , Pré-Escolar , Síndrome de Crigler-Najjar/genética , Síndrome de Crigler-Najjar/imunologia , Síndrome de Crigler-Najjar/patologia , Dependovirus/imunologia , Antagonistas de Aminoácidos Excitatórios/uso terapêutico , Feminino , Expressão Gênica , Glucuronosiltransferase/deficiência , Glucuronosiltransferase/imunologia , Células HEK293 , Humanos , Imunidade Inata , Imunização Passiva , Fígado/imunologia , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fenobarbital/uso terapêutico , Fototerapia/métodos , Plasmídeos/química , Plasmídeos/metabolismo , Transfecção
6.
Gastroenterology ; 152(4): 880-894.e6, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27939373

RESUMO

BACKGROUND & AIMS: Hepatocellular adenomas (HCAs) are benign liver tumors that can be assigned to molecular subtypes based on inactivating mutations in hepatocyte nuclear factor 1A, activating mutations in ß-catenin, or activation of inflammatory signaling pathways. We aimed to update the classification system for HCA and associate the subtypes with disease risk factors and complications. METHODS: We analyzed expression levels of 20 genes and sequenced exon regions of 8 genes (HNF1A, IL6ST, CTNNB1, FRK, STAT3, GNAS, JAK1, and TERT) in 607 samples of 533 HCAs from 411 patients, collected from 28 centers mainly in France from 2000 and 2014. We performed gene expression profile, RNA sequence, whole-exome and genome sequence, and immunohistochemical analyses of select samples. Molecular data were associated with risk factors, histopathology, bleeding, and malignant transformation. RESULTS: Symptomatic bleeding occurred in 14% of the patients (85% of cases were female, median age, 38 years); 7% of the nodules were borderline between HCA and hepatocellular carcinoma, and 3% of patients developed hepatocellular carcinoma from HCA. Based on molecular features, we classified HCA into 8 subgroups. One new subgroup, composed of previously unclassified HCA, represented 4% of HCAs overall and was associated with obesity and bleeding. These tumors were characterized by activation of sonic hedgehog signaling, due to focal deletions that fuse the promoter of INHBE with GLI1. Analysis of genetic heterogeneity among multiple HCAs, from different patients, revealed a molecular subtype field effect; multiple tumors had different mutations that deregulated similar pathways. Specific molecular subtypes of HCA associated with various HCA risk factors, including imbalances in estrogen or androgen hormones. Specific molecular subgroup of HCA with ß-catenin and sonic hedgehog activation associated with malignant transformation and bleeding, respectively. CONCLUSIONS: Using sequencing and gene expression analyses, we identified a subgroup of HCA characterized by fusion of the INHBE and GLI1 genes and activation of sonic hedgehog pathway. Molecular subtypes of HCAs associated with different patients' risk factors for HCA, disease progression, and pathology features of tumors. This classification system might be used to select treatment strategies for patients with HCA.


Assuntos
Adenoma/genética , Carcinoma Hepatocelular/genética , Subunidades beta de Inibinas/genética , Neoplasias Hepáticas/genética , Proteína GLI1 em Dedos de Zinco/genética , Adenoma/química , Adenoma/classificação , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma Hepatocelular/química , Carcinoma Hepatocelular/patologia , Transformação Celular Neoplásica/genética , Criança , Cromograninas/genética , Receptor gp130 de Citocina/genética , Feminino , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Fusão Gênica , Proteínas Hedgehog/metabolismo , Hemorragia/etiologia , Fator 1-alfa Nuclear de Hepatócito/genética , Humanos , Janus Quinase 2/genética , Neoplasias Hepáticas/química , Neoplasias Hepáticas/classificação , Masculino , Pessoa de Meia-Idade , Mutação , Proteínas de Neoplasias/genética , Proteínas Tirosina Quinases/genética , Fatores de Risco , Fator de Transcrição STAT3/genética , Análise de Sequência de RNA , Transdução de Sinais , Telomerase/genética , Transcriptoma , Adulto Jovem , beta Catenina/genética
7.
Hum Mol Genet ; 25(17): 3784-3797, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27436577

RESUMO

Glycogen storage disease type I (GSDI) is a rare metabolic disease due to glucose-6 phosphatase deficiency, characterized by fasting hypoglycemia. Patients also develop chronic kidney disease whose mechanisms are poorly understood. To decipher the process, we generated mice with a kidney-specific knockout of glucose-6 phosphatase (K.G6pc-/- mice) that exhibited the first signs of GSDI nephropathy after 6 months of G6pc deletion. We studied the natural course of renal deterioration in K.G6pc-/- mice for 18 months and observed the progressive deterioration of renal functions characterized by early tubular dysfunction and a later destruction of the glomerular filtration barrier. After 15 months, K.G6pc-/- mice developed tubular-glomerular fibrosis and podocyte injury, leading to the development of cysts and renal failure. On the basis of these findings, we were able to detect the development of cysts in 7 out of 32 GSDI patients, who developed advanced renal impairment. Of these 7 patients, 3 developed renal failure. In addition, no renal cysts were detected in six patients who showed early renal impairment. In conclusion, renal pathology in GSDI is characterized by progressive tubular dysfunction and the development of polycystic kidneys that probably leads to the development of irreversible renal failure in the late stages. Systematic observations of cyst development by kidney imaging should improve the evaluation of the disease's progression, independently of biochemical markers.


Assuntos
Barreira de Filtração Glomerular/patologia , Glucose-6-Fosfatase/genética , Doença de Depósito de Glicogênio Tipo I/complicações , Doenças Renais Císticas/etiologia , Insuficiência Renal/etiologia , Adolescente , Adulto , Animais , Criança , Pré-Escolar , Modelos Animais de Doenças , Progressão da Doença , Feminino , Técnicas de Inativação de Genes , Barreira de Filtração Glomerular/fisiopatologia , Doença de Depósito de Glicogênio Tipo I/genética , Doença de Depósito de Glicogênio Tipo I/fisiopatologia , Humanos , Lactente , Doenças Renais Císticas/patologia , Masculino , Camundongos , Pessoa de Meia-Idade , Insuficiência Renal/patologia , Adulto Jovem
8.
J Inherit Metab Dis ; 39(5): 697-704, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27106217

RESUMO

Glycogen storage disease type III (GSDIII) is a rare disorder of glycogenolysis due to AGL gene mutations, causing glycogen debranching enzyme deficiency and storage of limited dextrin. Patients with GSDIIIa show involvement of liver and cardiac/skeletal muscle, whereas GSDIIIb patients display only liver symptoms and signs. The International Study on Glycogen Storage Disease (ISGSDIII) is a descriptive retrospective, international, multi-centre cohort study of diagnosis, genotype, management, clinical course and outcome of 175 patients from 147 families (86 % GSDIIIa; 14 % GSDIIIb), with follow-up into adulthood in 91 patients. In total 58 AGL mutations (non-missense mutations were overrepresented and 21 novel mutations were observed) were identified in 76 families. GSDIII patients first presented before the age of 1.5 years, hepatomegaly was the most common presenting clinical sign. Dietary management was very diverse and included frequent meals, uncooked cornstarch and continuous gastric drip feeding. Chronic complications involved the liver (hepatic cirrhosis, adenoma(s), and/or hepatocellular carcinoma in 11 %), heart (cardiac involvement and cardiomyopathy, in 58 % and 15 %, respectively, generally presenting in early childhood), and muscle (pain in 34 %). Type 2 diabetes mellitus was diagnosed in eight out of 91 adult patients (9 %). In adult patients no significant correlation was detected between (non-) missense AGL genotypes and hepatic, cardiac or muscular complications. This study demonstrates heterogeneity in a large cohort of ageing GSDIII patients. An international GSD patient registry is warranted to prospectively define the clinical course, heterogeneity and the effect of different dietary interventions in patients with GSDIII.


Assuntos
Doença de Depósito de Glicogênio Tipo III/diagnóstico , Doença de Depósito de Glicogênio Tipo III/genética , Adolescente , Adulto , Criança , Pré-Escolar , Feminino , Seguimentos , Genótipo , Sistema da Enzima Desramificadora do Glicogênio/genética , Humanos , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Mutação/genética , Estudos Retrospectivos , Adulto Jovem
9.
PLoS One ; 9(2): e89713, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24586979

RESUMO

BACKGROUND: Several lines of evidence indicate that immature oocyte retrieval and subsequent in vitro maturation (IVM) without ovarian stimulation may be a reliable option in assisted reproductive technologies (ART). However, few outcome data are available for children born following this technique. OBJECTIVE: We assessed height and weight development of French children conceived after IVM. METHODS: All children conceived after IVM at Antoine Beclere Hospital (Clamart, France) and born between June 2003 and October 2008 (n = 38) were included in a prospective cohort study and compared with a control group of children conceived by ICSI without IVM, matched for maternal age, gestational age and singleton/twin pregnancies. Follow-up included clinical examination at one year and a questionnaire completed by parents when the children were two years old (97% follow-up rate). RESULTS: No statistical differences between IVM and control groups were found for boys. Mean weight, height and head circumference at birth were significantly greater for IVM than for ICSI girls (3.236 kg vs 2.701 kg (p = 0.03); 49 cm vs 47 cm (p = 0.01) and 34 cm vs 33 cm (p = 0.04), respectively). At one year, IVM girls remained heavier (mean weight 10.2 kg vs 8.6 kg (p = 0.001)) and taller (76 cm vs 73 cm (p = 0.03)), and there was a two-point difference in BMI between the two groups of girls (18 vs 16 (p = 0.01)). CONCLUSION: Our results in girls born after IVM should be interpreted with caution. It remains unclear whether the observed sexual dimorphism is due to IVM technology or to maternal characteristics such as underlying infertility in patients with polycystic ovary syndrome (PCOS). Further monitoring of the outcomes of these infants is required.


Assuntos
Desenvolvimento Infantil , Fertilização in vitro , Técnicas In Vitro , Resultado da Gravidez , Peso ao Nascer , Estatura , Peso Corporal , Cefalometria , Pré-Escolar , Transferência Embrionária , Feminino , Fertilização in vitro/métodos , Humanos , Lactente , Recém-Nascido , Masculino , Síndrome do Ovário Policístico , Gravidez , Gravidez de Gêmeos , Estudos Prospectivos , Injeções de Esperma Intracitoplásmicas
10.
Hum Gene Ther Methods ; 25(3): 181-6, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24593043

RESUMO

Helper-dependent adenoviral (HDAd) vectors are attractive for liver-directed gene therapy because they can drive sustained high levels of transgene expression without chronic toxicity. However, high vector doses are required to achieve efficient hepatic transduction by systemic delivery because of a nonlinear dose response. Unfortunately, such high doses result in systemic vector dissemination and dose-dependent acute toxicity with potential lethal consequences. We have previously shown in nonhuman primates that delivery of HDAd in surgically isolated livers resulted in a significantly higher hepatic transduction with reduced systemic vector dissemination compared with intravenous delivery and multiyear transgene expression. Encouraged by these data, we have now employed a surgical vector delivery method in the Gunn rat, an animal model for Crigler-Najjar syndrome. After vector delivery into the surgically isolated liver, we show phenotypic correction at the low and clinically relevant vector dose of 1 × 10(11) vp/kg. Correction of hyperbilirubinemia and increased glucuronidation of bilirubin in bile was achieved for up to 1 year after vector administration. Surgical delivery of the vector was well tolerated without signs of acute or chronic toxicity. This method of delivery could thereby be a safer alternative to liver transplantation for long-term treatment of Crigler-Najjar syndrome type I.


Assuntos
Adenoviridae/genética , Vetores Genéticos/metabolismo , Hiperbilirrubinemia/terapia , Animais , Bilirrubina/sangue , Síndrome de Crigler-Najjar/terapia , Terapia Genética , Vetores Genéticos/genética , Glucuronosiltransferase/genética , Humanos , Fígado/metabolismo , Fígado/cirurgia , Regiões Promotoras Genéticas , Ratos , Ratos Gunn , Transdução Genética
11.
J Hepatol ; 58(2): 350-7, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23046672

RESUMO

BACKGROUND & AIMS: Hepatocellular adenomas (HCA) are benign liver tumors mainly related to oral contraception and classified into 4 molecular subgroups: inflammatory (IHCA), HNF1A-inactivated (H-HCA), ß-catenin-activated (bHCA) or unclassified (UHCA). Glycogen storage disease type I (GSD) is a rare hereditary metabolic disease that predisposes to HCA development. The aim of our study was to characterize the molecular profile of GSD-associated HCA. METHODS: We characterized a series of 25 HCAs developed in 15 patients with GSD by gene expression and DNA sequence of HNF1A, CTNNB1, IL6ST, GNAS, and STAT3 genes. Moreover, we searched for glycolysis, gluconeogenesis, and fatty acid synthesis alterations in GSD non-tumor livers and compared our results to those observed in a series of sporadic H-HCA and various non-GSD liver samples. RESULTS: GSD adenomas were classified as IHCA (52%) mutated for IL6ST or GNAS, bHCA (28%) or UHCA (20%). In contrast, no HNF1A inactivation was observed, showing a different molecular subtype distribution in GSD-associated HCA from that observed in sporadic HCA (p = 0.0008). In non-tumor GSD liver samples, we identified glycolysis and fatty acid synthesis activation with gluconeogenesis repression. Interestingly, this gene expression profile was similar to that observed in sporadic H-HCA. CONCLUSIONS: Our study showed a particular molecular profile in GSD-related HCA characterized by a lack of HNF1A inactivation. This exclusion could be explained by similar metabolic defects observed with HNF1A inactivation and glucose-6-phosphatase deficiency. Inversely, the high frequency of ß-catenin mutations could be related to the increased frequency of malignant transformation in hepatocellular carcinoma.


Assuntos
Adenoma de Células Hepáticas/epidemiologia , Adenoma de Células Hepáticas/genética , Perfilação da Expressão Gênica , Doença de Depósito de Glicogênio Tipo I/epidemiologia , Doença de Depósito de Glicogênio Tipo I/genética , Neoplasias Hepáticas/epidemiologia , Neoplasias Hepáticas/genética , Adenoma de Células Hepáticas/metabolismo , Adolescente , Adulto , Cromograninas , Comorbidade , Receptor gp130 de Citocina/genética , Receptor gp130 de Citocina/metabolismo , Feminino , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Regulação Neoplásica da Expressão Gênica , Doença de Depósito de Glicogênio Tipo I/metabolismo , Fator 1-alfa Nuclear de Hepatócito/genética , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Humanos , Fígado/metabolismo , Fígado/patologia , Neoplasias Hepáticas/metabolismo , Masculino , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Adulto Jovem , beta Catenina/genética , beta Catenina/metabolismo
12.
J Hepatol ; 56(1): 184-91, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21835143

RESUMO

BACKGROUND & AIMS: Mosaic G-protein alpha-subunit (GNAS)-activating mutations are responsible for the McCune-Albright (MCA) syndrome. This oncogene that activates the adenylate cyclase is also mutated in various tumor types leading to the accumulation of cyclic-AMP. Identification of a hepatocellular adenoma (HCA) in two MCA patients led us to search for GNAS activation in benign and malignant hepatocellular carcinogenesis. METHODS: GNAS mutations were screened by sequencing 164 HCA, 245 hepatocellular carcinoma (HCC), and 17 fibrolamellar carcinomas. Tumors were characterized by quantitative RT-PCR, gene mutation screening and pathological reviewing. The consequences of wild type and mutant GNAS expression were analyzed in hepatocellular cell lines. RESULTS: A somatic GNAS-activating mutation was identified in 5 benign tumors and in 2 HCC. In benign tumors, GNAS mutations were exclusive from HNF1A, CTNNB1, and IL6ST mutations whereas one HCC demonstrated both CTNNB1 and GNAS mutations. Quantitative RT-PCR showed an activation of the IL-6 and interferon pathways in GNAS-mutated tumor tissues. Accordingly, pathological reviewing identified in GNAS-mutated tumors an inflammatory phenotype characterized by fibrosis and STAT3 activation. We further demonstrated in HCC cell lines that GNAS mutant expression induced inflammatory response and STAT3 activation. CONCLUSIONS: We identified for the first time the association between two rare diseases, MCA syndrome and HCA occurrence, but also that somatic GNAS-activating mutations in sporadic benign and malignant liver tumors are characterized by an inflammatory phenotype. These results showed a cross-talk between cyclic-AMP and JAK/STAT pathways in liver tumors and they reinforce the role of STAT3 activation in liver tumorigenesis.


Assuntos
Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Neoplasias Hepáticas/genética , Mutação , Fator de Transcrição STAT3/metabolismo , Adenoma de Células Hepáticas/genética , Adenoma de Células Hepáticas/metabolismo , Adenoma de Células Hepáticas/patologia , Adulto , Idoso , Sequência de Bases , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Cromograninas , Análise Mutacional de DNA , DNA de Neoplasias/genética , Feminino , Displasia Fibrosa Poliostótica/genética , Humanos , Neoplasias Hepáticas/classificação , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Transdução de Sinais
13.
Mol Genet Metab ; 104(1-2): 137-43, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21646031

RESUMO

Glycogen storage disease (GSD) due to a deficient hepatic phosphorylase system defines a genetically heterogeneous group of disorders that mainly manifests in children. We investigated 45 unrelated children in whom a liver GSD VI or IX was suspected on the basis of clinical symptoms including hepatomegaly, increased serum transaminases, postprandial lactatemia and/or mild fasting hypoglycemia. Liver phosphorylase and phosphorylase b kinase activities studied in peripheral blood cells allowed to suspect diagnosis in 37 cases but was uninformative in 5. Sequencing of liver phosphorylase genes was useful to establish an accurate diagnosis. Causative mutations were found either in the PYGL (11 patients), PHKA2 (26 patients), PHKG2 (three patients) or in the PHKB (three patients) genes. Eleven novel disease causative mutations, five missense (p.N188K, p.D228Y, p.P382L, p.R491H, p.L500R) and six truncating mutations (c.501_502ins361pb, c.528+2T>C, c.856-29_c.1518+614del, c.1620+1G>C, p.E703del and c.2313-1G>T) were identified in the PYGL gene. Seventeen novel disease causative mutations, ten missense (p.A42P, p.Q95R, p.G131D, p.G131V, p.Q134R, p.G187R, p.G300V, p.G300A, p.C326Y, p.W820G) and seven truncating (c.537+5G>A, p.G396DfsX28, p.Q404X, p.N653X, p.L855PfsX87, and two large deletions) were identified in the PHKA2 gene. Four novel truncating mutations (p.R168X, p.Q287X, p.I268PfsX12 and c.272-1G>C) were identified in the PHKG2 gene and three (c.573_577del, p.R364X, c.2427+3A>G) in the PHKB gene. Patients with PHKG2 mutations evolved towards cirrhosis. Molecular analysis of GSD VI or IX genes allows to confirm diagnosis suspected on the basis of enzymatic analysis and to establish diagnosis and avoid liver biopsy when enzymatic studies are not informative in blood cells.


Assuntos
Doença de Depósito de Glicogênio/sangue , Doença de Depósito de Glicogênio/diagnóstico , Fígado/enzimologia , Fígado/patologia , Fosforilase Quinase/deficiência , Fosforilases/deficiência , Pré-Escolar , Feminino , Estudos de Associação Genética , Doença de Depósito de Glicogênio/enzimologia , Doença de Depósito de Glicogênio/genética , Humanos , Lactente , Masculino , Mutação/genética , Fosforilase Quinase/genética , Fosforilases/genética
14.
Orphanet J Rare Dis ; 6: 27, 2011 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-21599942

RESUMO

Glucose-6-phosphatase deficiency (G6P deficiency), or glycogen storage disease type I (GSDI), is a group of inherited metabolic diseases, including types Ia and Ib, characterized by poor tolerance to fasting, growth retardation and hepatomegaly resulting from accumulation of glycogen and fat in the liver. Prevalence is unknown and annual incidence is around 1/100,000 births. GSDIa is the more frequent type, representing about 80% of GSDI patients. The disease commonly manifests, between the ages of 3 to 4 months by symptoms of hypoglycemia (tremors, seizures, cyanosis, apnea). Patients have poor tolerance to fasting, marked hepatomegaly, growth retardation (small stature and delayed puberty), generally improved by an appropriate diet, osteopenia and sometimes osteoporosis, full-cheeked round face, enlarged kydneys and platelet dysfunctions leading to frequent epistaxis. In addition, in GSDIb, neutropenia and neutrophil dysfunction are responsible for tendency towards infections, relapsing aphtous gingivostomatitis, and inflammatory bowel disease. Late complications are hepatic (adenomas with rare but possible transformation into hepatocarcinoma) and renal (glomerular hyperfiltration leading to proteinuria and sometimes to renal insufficiency). GSDI is caused by a dysfunction in the G6P system, a key step in the regulation of glycemia. The deficit concerns the catalytic subunit G6P-alpha (type Ia) which is restricted to expression in the liver, kidney and intestine, or the ubiquitously expressed G6P transporter (type Ib). Mutations in the genes G6PC (17q21) and SLC37A4 (11q23) respectively cause GSDIa and Ib. Many mutations have been identified in both genes,. Transmission is autosomal recessive. Diagnosis is based on clinical presentation, on abnormal basal values and absence of hyperglycemic response to glucagon. It can be confirmed by demonstrating a deficient activity of a G6P system component in a liver biopsy. To date, the diagnosis is most commonly confirmed by G6PC (GSDIa) or SLC37A4 (GSDIb) gene analysis, and the indications of liver biopsy to measure G6P activity are getting rarer and rarer. Differential diagnoses include the other GSDs, in particular type III (see this term). However, in GSDIII, glycemia and lactacidemia are high after a meal and low after a fast period (often with a later occurrence than that of type I). Primary liver tumors and Pepper syndrome (hepatic metastases of neuroblastoma) may be evoked but are easily ruled out through clinical and ultrasound data. Antenatal diagnosis is possible through molecular analysis of amniocytes or chorionic villous cells. Pre-implantatory genetic diagnosis may also be discussed. Genetic counseling should be offered to patients and their families. The dietary treatment aims at avoiding hypoglycemia (frequent meals, nocturnal enteral feeding through a nasogastric tube, and later oral addition of uncooked starch) and acidosis (restricted fructose and galactose intake). Liver transplantation, performed on the basis of poor metabolic control and/or hepatocarcinoma, corrects hypoglycemia, but renal involvement may continue to progress and neutropenia is not always corrected in type Ib. Kidney transplantation can be performed in case of severe renal insufficiency. Combined liver-kidney grafts have been performed in a few cases. Prognosis is usually good: late hepatic and renal complications may occur, however, with adapted management, patients have almost normal life span. DISEASE NAME AND SYNONYMS: Glucose-6-phosphatase deficiency or G6P deficiency or glycogen storage disease type I or GSDI or type I glycogenosis or Von Gierke disease or Hepatorenal glycogenosis.


Assuntos
Doença de Depósito de Glicogênio Tipo I/patologia , Testes Genéticos , Doença de Depósito de Glicogênio Tipo I/dietoterapia , Doença de Depósito de Glicogênio Tipo I/epidemiologia , Doença de Depósito de Glicogênio Tipo I/genética , Humanos , Mutação
15.
JIMD Rep ; 1: 97-106, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-23430835

RESUMO

The development of hepatocellular adenomas in the liver of patients with glycogen storage disease type I is a well-known complication of the disease. Surgical procedures and perioperative managements described so far have reported persistent and important morbidity. We report here a series of six patients (three males and three females) who underwent hepatic resection, and we propose a new hemostatic management protocol comprising glucose infusion, corticosteroids, desmopressin, and antifibrinolytic drugs, used to prevent efficaciously hepatic hemorrhage due to glycogen storage disease (GSD) platelet dysfunction.

16.
Gastroenterology ; 139(3): 999-1007, 1007.e1-2, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20546738

RESUMO

BACKGROUND & AIMS: Crigler-Najjar type 1 (CN-I) is an inherited liver disease caused by an absence of bilirubin-uridine 5'-diphosphate-glucuronosyltransferase (UGT1A1) activity. It results in life-threatening levels of unconjugated bilirubin, and therapeutic options are limited. We used adult Gunn rats (an animal model of the disease) to evaluate the efficiency of lentiviral-based gene therapy to express UGT1A1 in liver. METHODS: Gunn rats were given intraportal injections of VSVG-pseudotyped lentiviral vectors that encode UGT1A1 under the control of a liver-specific transthyretin promoter (mTTR.hUGT1A1); this vector does not contain target sequences for miR-142, a microRNA that is expressed specifically in hematopoietic cells. Rats were also injected with the vector mTTR.hUGT1A1.142T, which contains 4 copies of the miR-142 target sequences; its messenger RNA should be degraded in antigen-presenting cells. Bilirubinemia was monitored, and the presence of transduced hepatocytes was analyzed by quantitative polymerase chain reaction. Vector expression was tested in vitro in rat hematopoietic cells. RESULTS: In Gunn rats, bilirubin levels normalized 2 weeks after administration of mTTR.hUGT1A1. However, hyperbilirubinemia resumed 8 weeks after vector administration, concomitant with the induction of an immune response. In contrast, in rats injected with mTTR-UGT1A1.142T, bilirubin levels normalized for up to 6 months and transduced cells were not eliminated. CONCLUSIONS: Lentiviral vectors that express UGT1A1 reduce hyperbilirubinemia in immunocompetent Gunn rats for at least 6 months. The immune response against virally expressed UGT1A1 can be circumvented by inclusion of miR-142 target sequences, which reduce vector expression in antigen-presenting cells. This lentiviral-based gene therapy approach might be developed to treat patients with CN-I.


Assuntos
Síndrome de Crigler-Najjar/terapia , Terapia Genética/métodos , Vetores Genéticos , Glucuronosiltransferase/genética , Lentivirus/genética , Fígado/enzimologia , MicroRNAs/metabolismo , Animais , Anticorpos/sangue , Células Apresentadoras de Antígenos/imunologia , Bilirrubina/sangue , Biomarcadores/sangue , Síndrome de Crigler-Najjar/enzimologia , Síndrome de Crigler-Najjar/genética , Síndrome de Crigler-Najjar/imunologia , Modelos Animais de Doenças , Glucuronosiltransferase/biossíntese , Glucuronosiltransferase/imunologia , Células HeLa , Humanos , Masculino , Pré-Albumina/genética , Regiões Promotoras Genéticas , Estabilidade de RNA , RNA Mensageiro/metabolismo , Ratos , Ratos Gunn , Fatores de Tempo , Transdução Genética
17.
Fetal Diagn Ther ; 21(5): 428-32, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16912492

RESUMO

OBJECTIVES: CVS is the earliest procedure for cytogenetic analysis but the quality of metaphases obtained does not allow the characterization of subtle chromosomal anomalies. We report the application interphase fluorescence in situ hybridization for the rapid prenatal diagnosis of a subtle structural chromosome anomaly in trophoblast cells. METHODS AND RESULTS: The foetus was karyotyped because of a paternal complex chromosomal anomaly 46,XY,inv(2)(q14.3q35),ins(10;2)(q25;q36.1q36.1). Fluorescence in situ hybridization analyses were performed on interphase nuclei and metaphase chromosomes from uncultured chorionic villi using bacterial artificial chromosomes specific for the 2q chromosomal region. Direct conventional cytogenetics showed an apparently normal male karyotype, whereas fluorescence in situ hybridization analysis showed a deletion of the chromosomal region 2q36.1 and a paracentric inversion of the chromosome 2q leading to a partial monosomy 2q36.1. CONCLUSION: This strategy allowed us to offer an early and rapid chromosomal analysis for this couple leading to a better management of the pregnancy. This report demonstrates that interphase fluorescence in situ hybridization can be used in direct CVS for a rapid and early prenatal diagnosis of complex chromosomal rearrangements.


Assuntos
Cromossomos Humanos Par 2/genética , Monossomia/diagnóstico , Diagnóstico Pré-Natal/métodos , Trofoblastos/ultraestrutura , Pré-Escolar , Amostra da Vilosidade Coriônica , Bandeamento Cromossômico , Inversão Cromossômica/genética , Feminino , Deleção de Genes , Idade Gestacional , Humanos , Hibridização in Situ Fluorescente , Gravidez
18.
J Pediatr Gastroenterol Nutr ; 37(5): 566-70, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14581798

RESUMO

OBJECTIVES: Glycogen storage disease type Ia (GSD-Ia), a congenital deficiency of hepatic glucose-6-phosphatase activity, is often associated with hyperproteinemia. To document the mechanism of hyperproteinemia, the proteins of the hemostatic system were analyzed according to their site of synthesis: hepatocyte, endothelial cell, or both. The role of inflammation was investigated by the measurement of tumor necrosis factor alpha (TNF-alpha) and interleukin-6 (IL-6) levels in plasma. METHODS: Twenty-seven patients with GSD-Ia were evaluated, as were 14 patients with other types of GSD and 30 healthy control subjects. Of the 41 patients with GSD, 15 also had hepatic adenoma (14 patients with GSD-Ia and 1 with GSD type III). RESULTS: In patients with GSD-Ia, there was a two-fold increase in all hepatocyte-synthesized proteins (i.e., factor VII, protein C, C4b binding protein) compared with control subjects and patients with other types of GSD. The proteins with mixed endothelial and hepatocyte origin (i.e., antithrombin and protein S) also were significantly increased but to a lesser extent. In contrast, the mean concentration of von Willebrand factor, which is exclusively synthesized in endothelial cells, was normal, as was the concentration of TNF-alpha and IL-6. CONCLUSIONS: These results suggest that the hyperproteinemia of GSD-Ia (including hemostatic proteins) is attributable to hepatocyte dysfunction and not related to an inflammatory process.


Assuntos
Proteínas Sanguíneas/análise , Proteínas Inativadoras do Complemento , Doença de Depósito de Glicogênio Tipo I/sangue , Hemostasia , Inflamação/sangue , Adolescente , Adulto , Antitrombinas/análise , Criança , Pré-Escolar , Células Endoteliais/metabolismo , Fator VII/análise , Glicoproteínas/sangue , Humanos , Interleucina-6/análise , Fígado/metabolismo , Inibidor 1 de Ativador de Plasminogênio/sangue , Proteína C/análise , Proteína S/análise , Triglicerídeos/sangue , Fator de Necrose Tumoral alfa/análise , Fator de von Willebrand/análise
19.
Prenat Diagn ; 22(10): 914-6, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12378576

RESUMO

Crigler-Najjar syndrome type I (CN-I) is a rare and severe inherited disorder of bilirubin metabolism, caused by the total deficiency of bilirubin-UDP-glucuronosyltransferase (UGT) activity. Enzymatic diagnosis cannot be performed in chorionic villi or amniocytes as UGT is not active in these tissues. The cloning of the UGT1 gene and the identification of disease-causing mutations have led to the possibility of performing DNA-based diagnosis. Here we report DNA-based prenatal diagnosis of CN-I in two Tunisian families in whom CN-I patients were diagnosed. As we had previously shown that CN-I was, in Tunisia, associated with homozygosity for the Q357R mutation within the UGT1 gene, we were able to detect this mutation in both families and to show that it was easily recognized by single-strand conformation polymorphism (SSCP) analysis. In both cases, SSCP analysis of fetal DNA showed that the fetus was heterozygous for the Q357R mutation. In one family, the pregnancy was carried to term and a healthy baby was born, whereas, in the other family, the pregnancy is still continuing. Thus the prenatal diagnosis of CN-I is possible, provided disease-causing mutations have been identified. SSCP analysis of DNA prepared either from amniocytes or from chorionic villi is a simple, reliable and fast method for prenatal diagnosis.


Assuntos
Síndrome de Crigler-Najjar/diagnóstico , Síndrome de Crigler-Najjar/genética , Glucuronosiltransferase/genética , Polimorfismo Conformacional de Fita Simples , Diagnóstico Pré-Natal/métodos , Amostra da Vilosidade Coriônica , Feminino , Heterozigoto , Homozigoto , Humanos , Proteínas de Transporte de Monossacarídeos/genética , Mutação , Gravidez , Tunísia
20.
Eur J Pediatr ; 161 Suppl 1: S20-34, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12373567

RESUMO

UNLABELLED: Glycogen storage disease type I (GSD I) is a relatively rare metabolic disease and therefore, no metabolic centre has experience of large numbers of patients. To document outcome, to develop guidelines about (long-term) management and follow-up, and to develop therapeutic strategies, the collaborative European Study on GSD I (ESGSD I) was initiated. This paper is a descriptive analysis of data obtained from the retrospective part of the ESGSD I. Included were 231 GSD Ia and 57 GSD Ib patients. Median age of data collection was 10.4 years (range 0.4-45.4 years) for Ia and 7.1 years (0.4-30.6 years) for Ib patients. Data on dietary treatment, pharmacological treatment, and outcome including mental development, hyperlipidaemia and its complications, hyperuricaemia and its complications, bleeding tendency, anaemia, osteopenia, hepatomegaly, liver adenomas and carcinomas, progressive renal disease, height and adult height, pubertal development and bone maturation, school type, employment, and pregnancies are presented. Data on neutropenia, neutrophil dysfunction, infections, inflammatory bowel disease, and the use of granulocyte colony-stimulating factor are presented elsewhere (Visser et al. 2000, J Pediatr 137:187-191; Visser et al. 2002, Eur J Pediatr DOI 10.1007/s00431-002-1010-0). CONCLUSION: there is still wide variation in methods of dietary and pharmacological treatment of glycogen storage disease type I. Intensive dietary treatment will improve, but not correct completely, clinical and biochemical status and fewer patients will die as a direct consequence of acute metabolic derangement. With ageing, more and more complications will develop of which progressive renal disease and the complications related to liver adenomas are likely to be two major causes of morbidity and mortality.


Assuntos
Doença de Depósito de Glicogênio Tipo I , Adenoma/etiologia , Estatura , Criança , Pré-Escolar , Feminino , Doença de Depósito de Glicogênio Tipo I/complicações , Doença de Depósito de Glicogênio Tipo I/diagnóstico , Doença de Depósito de Glicogênio Tipo I/fisiopatologia , Doença de Depósito de Glicogênio Tipo I/terapia , Humanos , Lactente , Recém-Nascido , Neoplasias Hepáticas/etiologia , Masculino , Estudos Retrospectivos , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA