Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Artigo em Inglês | MEDLINE | ID: mdl-32518610

RESUMO

PURPOSE: In order to further understand genetically predisposing factors of gastric cancer, a retrospective study on 107 patients with gastric cancer was conducted. The family history of cancer cases was registered, in search of associations between gastric cancer and other cancer types. MATERIALS AND METHODS: Within Stockholm County in Sweden, all patients previously diagnosed with gastric cancer and still alive were invited to participate in the study. Patients were asked to complete a questionnaire about their gastric cancer diagnosis and if any cancers had occurred in their family. A blood sample for DNA extraction was collected. The proportions of different cancer types in the relatives of the patients were compared to the general Swedish population in 1970 and 2010. RESULTS: Among first- and second-degree relatives to the index patients with gastric cancer, the frequency of uterine cancer as well as gastric cancer was significantly overrepresented compared to the general population in Sweden. The frequency of breast cancer was significantly lower. CONCLUSIONS: There seems to be an increased risk of both gastric cancer and uterine cancer in the families of gastric cancer survivors, indicating a possible hereditary connection between these two cancer types.

2.
J Cell Mol Med ; 16(1): 129-41, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21342435

RESUMO

The syntaxin 11 (STX11) gene is mutated in a proportion of patients with familial haemophagocytic lymphohistiocytosis (FHL) and exocytosis of cytotoxic granules is impaired in STX11-deficient NK cells. However, the subcellular localization, regulation of expression and molecular function of STX11 in NK cells and other cytotoxic lymphocytes remain unknown. Here we demonstrate that STX11 expression is strictly controlled by several mechanisms in a cell-type-specific manner and that the enzymatic activity of the proteasome is required for STX11 expression in NK cells. In resting NKL cells, STX11 was localized in the cation-dependent mannose-6-phosphate receptor (CD-M6PR)-containing compartment, which was clearly distinct from cytotoxic granules or Rab27a-expressing vesicles. These subcellular structures appeared to fuse at the contact area with NK-sensitive target cells as demonstrated by partial colocalization of STX11 with perforin and Rab27a. Although STX11-deficent allo-specific cytotoxic T-lymphocytes efficiently lysed target cells and released cytotoxic granules, they exhibited a significantly lower extent of spontaneous association of perforin with Rab27a as compared with STX11-expressing T cells. Thus, our results suggest that STX11 promotes the fusion of Rab27a-expressing vesicles with cytotoxic granules and reveal an additional level of complexity in the spatial/temporal segregation of subcellular structures participating in the process of granule-mediated cytotoxicity.


Assuntos
Grânulos Citoplasmáticos/metabolismo , Sinapses Imunológicas/imunologia , Células Matadoras Naturais/citologia , Células Matadoras Naturais/imunologia , Proteínas Qa-SNARE/metabolismo , Linhagem Celular , Humanos , Linfo-Histiocitose Hemofagocítica/genética , Linfo-Histiocitose Hemofagocítica/fisiopatologia , Perforina/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Qa-SNARE/genética , Frações Subcelulares/metabolismo , Linfócitos T Citotóxicos/imunologia , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas rab27 de Ligação ao GTP
3.
Pediatr Blood Cancer ; 54(4): 563-72, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19953648

RESUMO

BACKGROUND: Griscelli syndrome type 2 (GS2) is an autosomal-recessive immunodeficiency caused by mutations in RAB27A, clinically characterized by partial albinism and haemophagocytic lymphohistocytosis (HLH). We evaluated the frequency of RAB27A mutations in 21 unrelated patients with haemophagocytic syndromes without mutations in familial HLH (FHL) causing genes or an established diagnosis of GS2. In addition, we report three patients with known GS2. Moreover, neurological involvement and RAB27A mutations in previously published patients with genetically verified GS2 are reviewed. PROCEDURE: Mutation analysis of RAB27A was performed by direct DNA sequencing. NK cell activity was evaluated and microscopy of the hair was performed to confirm the diagnosis. RESULTS: RAB27A mutations were found in 1 of the 21 families. This Swedish family had three affected children with heterozygous compound mutations consisting of a novel splice error mutation, [c.239G>C], and a nonsense mutation, [c.550C>T], p.R184X. The three additional children all carried homozygous RAB27A mutations, one of which is a novel splice error mutation, [c.240-2A>C]. Of note, five of the six patients displayed neurological symptoms, while three out of six patients displayed NK cell activity within normal reference values, albeit low. A literature review revealed that 67% of GS2 patients have been reported with neurological manifestations. CONCLUSIONS: Identification of RAB27A mutations can facilitate prompt diagnosis and treatment, and aid genetic counselling and prenatal diagnosis. Since five of six patients studied herein initially were diagnosed as having FHL, we conclude that the diagnosis of GS2 may be overlooked, particularly in fair-haired patients with haemophagocytic syndromes.


Assuntos
Síndromes de Imunodeficiência/diagnóstico , Síndromes de Imunodeficiência/genética , Linfo-Histiocitose Hemofagocítica/genética , Proteínas rab de Ligação ao GTP/genética , Anormalidades Múltiplas/genética , Adolescente , Albinismo/genética , Criança , Análise Mutacional de DNA , Feminino , Cabelo/ultraestrutura , Humanos , Lactente , Células Matadoras Naturais/imunologia , Masculino , Microscopia Eletrônica de Transmissão , Mutação , Linhagem , Proteínas rab27 de Ligação ao GTP
4.
Pediatr Blood Cancer ; 52(4): 527-9, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19058215

RESUMO

A patient with previously unrecognized X-linked chronic granulomatous disease (X-CGD) died of multi-organ failure, secondary to ongoing infection and hemophagocytic lymphohistiocytosis (HLH). Post mortem histological investigations were compatible with X-CGD, and a CYBB gene mutation was confirmed. No homozygous mutations in the genes encoding perforin (PRF1), MUNC 13-4 or syntaxin-11 (STX11) were found; however, there was a heterozygous alteration c.1471G>A in the PRF1 gene causing a p.Asp491Asn substitution. Although this substitution has not been reported to cause primary or secondary HLH, we speculate that it may have made the patient more susceptible for HLH under the circumstances of ongoing infection associated with X-CGD.


Assuntos
Doença Granulomatosa Crônica/complicações , Doença Granulomatosa Crônica/genética , Linfo-Histiocitose Hemofagocítica/complicações , Linfo-Histiocitose Hemofagocítica/genética , Proteínas Citotóxicas Formadoras de Poros/genética , Pré-Escolar , Evolução Fatal , Feminino , Doença Granulomatosa Crônica/fisiopatologia , Humanos , Linfo-Histiocitose Hemofagocítica/fisiopatologia , Masculino , Glicoproteínas de Membrana/genética , NADPH Oxidase 2 , NADPH Oxidases/genética , Linhagem , Perforina , Polimorfismo Genético
5.
Br J Haematol ; 143(1): 75-83, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18710388

RESUMO

Familial hemophagocytic lymphohistiocytosis (FHL) is a rare autosomal recessive lethal condition characterized by fever, cytopenia, hepatosplenomegaly and hemophagocytosis. The hallmark of FHL is defect apoptosis triggering and lymphocyte cellular cytotoxicity. Thus far three disease-causing genes (PRF1, UNC13D, STX11) have been identified. We performed a genotype-phenotype study in a large, multi-ethnic cohort of 76 FHL patients originating from 65 unrelated families. Biallelic mutations in PRF1, UNC13D and STX11 were demonstrated in 13/74 (18%), 6/61 (10%) and 14/70 (20%) patients, respectively. In 27/60 (45%) patients analyzed for all three genes, no molecular diagnosis was established. STX11 mutations were most common in Turkish families (7/28, 25%), whereas in Middle East families, PRF1 mutations were most frequent (6/13, 46%). No biallelic mutation was identified in most families of Nordic origin (13/14, 93%). Patients carrying PRF1 mutations had higher risk of early onset (age <6 months) compared to patients carrying STX11 mutations [adjusted odds ratio 8.23 (95% confidence interval [CI] = 1.20-56.40), P = 0.032]. Moreover, patients without identified mutations had increased risk of pathological cerebrospinal fluid (CSF) at diagnosis compared to patients with STX11 mutations [adjusted odds ratio 26.37 (CI = 1.90-366.82), P = 0.015]. These results indicate that the disease-causing mutations in FHL have different phenotypes with regard to ethnic origin, age at onset, and pathological CSF at diagnosis.


Assuntos
Linfo-Histiocitose Hemofagocítica/genética , Proteínas de Membrana/genética , Mutação , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Qa-SNARE/genética , Idade de Início , Pré-Escolar , Estudos de Coortes , Feminino , Genótipo , Humanos , Lactente , Recém-Nascido , Modelos Logísticos , Linfo-Histiocitose Hemofagocítica/líquido cefalorraquidiano , Linfo-Histiocitose Hemofagocítica/etnologia , Masculino , Razão de Chances , Omã/etnologia , Perforina , Fenótipo , Medição de Risco/métodos , Suécia/etnologia , Turquia/etnologia
6.
Pediatr Blood Cancer ; 50(5): 1067-9, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18000860

RESUMO

In the present study, DNA sequencing of the genes SRGN, ARF6, AP3B1, and SH2D1A was performed in a well defined cohort of 18 families with familial hemophagocytic lymphohistiocytosis (FHL). A heterozygous nucleotide change (C > T) in the 3'untranslated region of the SRGN gene and a monoallelic 3-base pair deletion (c.2409_2411delGAA) in exon 21 of the AP3B1 gene were observed in two different families. Additionally, two novel polymorphisms, one in intron 17 of AP3B1 and another in intron 2 of SH2D1A were identified. We conclude that mutations in SRGN, ARF6, and AP3B1 are not likely to be common in patients fulfilling the FHL criteria.


Assuntos
Fatores de Ribosilação do ADP/genética , Complexo 3 de Proteínas Adaptadoras/genética , Subunidades beta do Complexo de Proteínas Adaptadoras/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Linfo-Histiocitose Hemofagocítica/genética , Polimorfismo Genético/genética , Proteoglicanas/genética , Proteínas de Transporte Vesicular/genética , Regiões 3' não Traduzidas/genética , Fator 6 de Ribosilação do ADP , Criança , Pré-Escolar , Estudos de Coortes , Análise Mutacional de DNA , Éxons/genética , Feminino , Humanos , Lactente , Masculino , Mutação/genética , Proteína Associada à Molécula de Sinalização da Ativação Linfocitária
7.
Leuk Lymphoma ; 57(9): 2161-70, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27090575

RESUMO

To identify copy number alterations (CNAs) in pediatric B-cell precursor acute lymphoblastic leukemia (BCP ALL), array comparative genomic hybridization was performed on 50 cases; detected CNAs were validated in a cohort of 191 cases analyzed by single nucleotide polymorphism arrays. Apart from CNAs involving leukemia-associated genes, recurrent deletions targeting genes not previously implicated in BCP ALL, e.g. INIP, IRF1 and PDE4B, were identified. Deletions of the DNA repair gene INIP were exclusively found in cases with t(12;21), and deletions of SH2B3 were associated with intrachromosomal amplification of chromosome 21 (p < 0.001). A majority of BTLA deletions (7/11; 64%) affected samples with gain of 21q chromosome material, suggesting that BTLA deletions are associated with both germline and somatic gain of chromosome 21. In cases without known risk-associated cytogenetic markers, CNAs associated with adverse prognosis were identified in 50% (10/20), indicating that a majority of these cases could be assigned to distinct genetic subtypes.


Assuntos
Deleção de Genes , Dosagem de Genes , Regulação Neoplásica da Expressão Gênica , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patologia , Adolescente , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Transformação Celular Neoplásica/genética , Criança , Pré-Escolar , Aberrações Cromossômicas , Hibridização Genômica Comparativa , Biologia Computacional/métodos , Variações do Número de Cópias de DNA , Progressão da Doença , Epigênese Genética , Feminino , Perfilação da Expressão Gênica , Humanos , Lactente , Recém-Nascido , Masculino , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/mortalidade , Recidiva
8.
Nat Genet ; 48(11): 1359-1369, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27668656

RESUMO

TSHZ3, which encodes a zinc-finger transcription factor, was recently positioned as a hub gene in a module of the genes with the highest expression in the developing human neocortex, but its functions remained unknown. Here we identify TSHZ3 as the critical region for a syndrome associated with heterozygous deletions at 19q12-q13.11, which includes autism spectrum disorder (ASD). In Tshz3-null mice, differentially expressed genes include layer-specific markers of cerebral cortical projection neurons (CPNs), and the human orthologs of these genes are strongly associated with ASD. Furthermore, mice heterozygous for Tshz3 show functional changes at synapses established by CPNs and exhibit core ASD-like behavioral abnormalities. These findings highlight essential roles for Tshz3 in CPN development and function, whose alterations can account for ASD in the newly defined TSHZ3 deletion syndrome.


Assuntos
Transtorno do Espectro Autista/genética , Proteínas de Homeodomínio/genética , Neocórtex/patologia , Neurônios/patologia , Fatores de Transcrição/genética , Animais , Transtorno do Espectro Autista/patologia , Deleção Cromossômica , Cromossomos Humanos Par 19 , Feminino , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Haploinsuficiência , Heterozigoto , Humanos , Masculino , Camundongos , Camundongos Endogâmicos CBA , Neocórtex/embriologia , Neurogênese/genética , Sinapses/genética
10.
Acta Paediatr ; 96(11): 1703-6, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17888050

RESUMO

Hemophagocytic lymphohistiocytosis (HLH) is a rare condition with high mortality. We report an extremely premature girl, born in the 24th gestational week (BW 732 g), that during her second month developed a severe HLH subsequent to a Serratia marcescens septicemia, with hepatosplenomegaly, cytopenias, hyperbilirubinemia (mostly conjugated, total bilirubin 916 mumol/L), hypertriglyceridemia, hypofibrinogenemia, hyperferritinemia (21266 mug/L), and elevated sIL-2 receptor levels. Genetic analysis revealed no PRF1, STX11 or UNC13D gene mutations. Treatment was provided according to the HLH-2004 protocol with etoposide, dexamethasone, and immunoglobulin, but no cyclosporin because of immature kidneys. She recovered fully from the HLH but developed a severe retinopathy as well as green teeth secondary to the hyperbilirubinemia. We conclude that secondary, bacteria-associated HLH can develop in premature infants, and that HLH can be treated with cytotoxic therapy also in premature infants. It is important to be aware of HLH in premature infants, since it is treatable.


Assuntos
Linfo-Histiocitose Hemofagocítica/diagnóstico , Linfo-Histiocitose Hemofagocítica/tratamento farmacológico , Sepse/diagnóstico , Sepse/tratamento farmacológico , Infecções por Serratia/diagnóstico , Infecções por Serratia/tratamento farmacológico , Corticosteroides/administração & dosagem , Citotoxinas/administração & dosagem , Citotoxinas/uso terapêutico , Dexametasona/administração & dosagem , Quimioterapia Combinada , Etoposídeo/administração & dosagem , Feminino , Humanos , Hiperbilirrubinemia/diagnóstico , Imunoglobulinas/administração & dosagem , Recém-Nascido de Peso Extremamente Baixo ao Nascer , Recém-Nascido , Recém-Nascido Prematuro , Linfo-Histiocitose Hemofagocítica/microbiologia , Retinopatia da Prematuridade , Sepse/complicações , Sepse/microbiologia , Infecções por Serratia/complicações , Serratia marcescens
11.
Blood ; 110(6): 1906-15, 2007 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-17525286

RESUMO

Familial hemophagocytic lymphohistiocytosis (FHL) is typically an early onset, fatal disease characterized by a sepsislike illness with cytopenia, hepatosplenomegaly, and deficient lymphocyte cytotoxicity. Disease-causing mutations have been identified in genes encoding perforin (PRF1/FHL2), Munc13-4 (UNC13D/FHL3), and syntaxin-11 (STX11/FHL4). In contrast to mutations leading to loss of perforin and Munc13-4 function, it is unclear how syntaxin-11 loss-of-function mutations contribute to disease. We show here that freshly isolated, resting natural killer (NK) cells and CD8(+) T cells express syntaxin-11. In infants, NK cells are the predominant perforin-containing cell type. NK cells from FHL4 patients fail to degranulate when encountering susceptible target cells. Unexpectedly, IL-2 stimulation partially restores degranulation and cytotoxicity by NK cells, which could explain the less severe disease progression observed in FHL4 patients, compared with FHL2 and FHL3 patients. Since the effector T-cell compartment is still immature in infants, our data suggest that the observed defect in NK-cell degranulation may contribute to the pathophysiology of FHL, that evaluation of NK-cell degranulation in suspected FHL patients may facilitate diagnosis, and that these new insights may offer novel therapeutic possibilities.


Assuntos
Regulação da Expressão Gênica , Células Matadoras Naturais/citologia , Linfócitos/citologia , Linfo-Histiocitose Hemofagocítica/genética , Proteínas Qa-SNARE/genética , Adulto , Western Blotting , Proliferação de Células , Pré-Escolar , Citocinas/metabolismo , Feminino , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Lactente , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Células Matadoras Naturais/metabolismo , Proteínas com Domínio LIM , Proteínas com Homeodomínio LIM , Subpopulações de Linfócitos , Linfócitos/metabolismo , Linfo-Histiocitose Hemofagocítica/metabolismo , Linfo-Histiocitose Hemofagocítica/patologia , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Mutação/genética , Perforina , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Proteínas Qa-SNARE/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa