Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
3.
Orphanet J Rare Dis ; 17(1): 421, 2022 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-36461014

RESUMO

BACKGROUND: ENPP1 Deficiency-caused by biallelic variants in ENPP1-leads to widespread arterial calcification in early life (Generalized Arterial Calcification of Infancy, GACI) or hypophosphatemic rickets in later life (Autosomal Recessive Hypophosphatemic Rickets type 2, ARHR2). A prior study using the Exome Aggregation Consortium (ExAC)-a database of exomes obtained from approximately 60,000 individuals-estimated the genetic prevalence at approximately 1 in 200,000 pregnancies. METHODS: We estimated the genetic prevalence of ENPP1 Deficiency by evaluating allele frequencies from a population database, assuming Hardy-Weinberg equilibrium. This estimate benefitted from a comprehensive literature review using Mastermind ( https://mastermind.genomenon.com/ ), which uncovered additional variants and supporting evidence, a larger population database with approximately 140,000 individuals, and improved interpretation of variants as per current clinical guidelines. RESULTS: We estimate a genetic prevalence of approximately 1 in 64,000 pregnancies, thus more than tripling the prior estimate. In addition, the carrier frequency of ENPP1 variants was found to be highest in East Asian populations, albeit based on a small sample. CONCLUSION: These results indicate that a significant number of patients with ENPP1 Deficiency remain undiagnosed. Efforts to increase disease awareness as well as expand genetic testing, particularly in non-European populations are warranted, especially now that clinical trials for enzyme replacement therapy, which proved successful in animal models, are underway.


Assuntos
Raquitismo Hipofosfatêmico Familiar , Raquitismo Hipofosfatêmico , Animais , Feminino , Gravidez , Humanos , Prevalência , Povo Asiático , Bases de Dados Factuais
4.
Hum Mutat ; 43(12): 1673-1705, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36150100

RESUMO

Loss-of-function variants in the ectonucleotide pyrophosphatase/phosphodiesterase family member 1 (ENPP1) cause ENPP1 Deficiency, a rare disorder characterized by pathological calcification, neointimal proliferation, and impaired bone mineralization. The consequence of ENPP1 Deficiency is a broad range of age dependent symptoms and morbidities including cardiovascular complications and 50% mortality in infants, autosomal recessive hypophosphatemic rickets type 2 (ARHR2) in children, and joint pain, osteomalacia and enthesopathies in adults. Recent research continues to add to the growing clinical presentation profile as well as expanding the role of ENPP1 itself. Here we review the current knowledge on the spectrum of clinical and genetic findings of ENPP1 Deficiency reported in patients diagnosed with GACI or ARHR2 phenotypes using a comprehensive database of known ENPP1 variants with associated clinical data. A total of 108 genotypes were identified from 154 patients. Of the 109 ENPP1 variants reviewed, 72.5% were demonstrably disease-causing, a threefold increase in pathogenic/likely pathogenic variants over other databases. There is substantial heterogeneity in disease severity, even among patients with the same variant. The approach to creating a continuously curated database of ENPP1 variants accessible to clinicians is necessary to increase the diagnostic yield of clinical genetic testing and accelerate diagnosis of ENPP1 Deficiency.


Assuntos
Raquitismo Hipofosfatêmico Familiar , Diester Fosfórico Hidrolases , Pirofosfatases , Humanos , Raquitismo Hipofosfatêmico Familiar/genética , Mutação , Diester Fosfórico Hidrolases/genética , Pirofosfatases/genética
5.
Am J Hum Genet ; 109(9): 1605-1619, 2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-36007526

RESUMO

Newborn screening (NBS) dramatically improves outcomes in severe childhood disorders by treatment before symptom onset. In many genetic diseases, however, outcomes remain poor because NBS has lagged behind drug development. Rapid whole-genome sequencing (rWGS) is attractive for comprehensive NBS because it concomitantly examines almost all genetic diseases and is gaining acceptance for genetic disease diagnosis in ill newborns. We describe prototypic methods for scalable, parentally consented, feedback-informed NBS and diagnosis of genetic diseases by rWGS and virtual, acute management guidance (NBS-rWGS). Using established criteria and the Delphi method, we reviewed 457 genetic diseases for NBS-rWGS, retaining 388 (85%) with effective treatments. Simulated NBS-rWGS in 454,707 UK Biobank subjects with 29,865 pathogenic or likely pathogenic variants associated with 388 disorders had a true negative rate (specificity) of 99.7% following root cause analysis. In 2,208 critically ill children with suspected genetic disorders and 2,168 of their parents, simulated NBS-rWGS for 388 disorders identified 104 (87%) of 119 diagnoses previously made by rWGS and 15 findings not previously reported (NBS-rWGS negative predictive value 99.6%, true positive rate [sensitivity] 88.8%). Retrospective NBS-rWGS diagnosed 15 children with disorders that had been undetected by conventional NBS. In 43 of the 104 children, had NBS-rWGS-based interventions been started on day of life 5, the Delphi consensus was that symptoms could have been avoided completely in seven critically ill children, mostly in 21, and partially in 13. We invite groups worldwide to refine these NBS-rWGS conditions and join us to prospectively examine clinical utility and cost effectiveness.


Assuntos
Triagem Neonatal , Medicina de Precisão , Criança , Estado Terminal , Testes Genéticos/métodos , Humanos , Recém-Nascido , Triagem Neonatal/métodos , Estudos Retrospectivos
6.
Hum Mutat ; 43(2): 143-157, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34806794

RESUMO

X-linked hypophosphatemia (XLH), the most common form of hereditary hypophosphatemia, is caused by disrupting variants in the PHEX gene, located on the X chromosome. XLH is inherited in an X-linked pattern with complete penetrance observed for both males and females. Patients experience lifelong symptoms resulting from chronic hypophosphatemia, including impaired bone mineralization, skeletal deformities, growth retardation, and diminished quality of life. This chronic condition requires life-long management with disease-specific therapies, which can improve patient outcomes especially when initiated early in life. To centralize and disseminate PHEX variant information, we have established a new PHEX gene locus-specific database, PHEX LSDB. As of April 30, 2021, 870 unique PHEX variants, compiled from an older database of PHEX variants, a comprehensive literature search, a sponsored genetic testing program, and XLH clinical trials, are represented in the PHEX LSDB. This resource is publicly available on an interactive, searchable website (https://www.rarediseasegenes.com/), which includes a table of variants and associated data, graphical/tabular outputs of genotype-phenotype analyses, and an online submission form for reporting new PHEX variants. The database will be updated regularly with new variants submitted on the website, identified in the published literature, or shared from genetic testing programs.


Assuntos
Bases de Dados Genéticas , Raquitismo Hipofosfatêmico Familiar , Doenças Genéticas Ligadas ao Cromossomo X , Hipofosfatemia , Endopeptidase Neutra Reguladora de Fosfato PHEX , Raquitismo Hipofosfatêmico Familiar/genética , Feminino , Doenças Genéticas Ligadas ao Cromossomo X/genética , Humanos , Hipofosfatemia/genética , Masculino , Endopeptidase Neutra Reguladora de Fosfato PHEX/genética , Qualidade de Vida
7.
Front Genet ; 11: 577152, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33281875

RESUMO

Design and interpretation of genome sequencing assays in clinical diagnostics and research labs is complicated by an inability to identify information from the medical literature and related databases quickly, comprehensively and reproducibly. This challenge is compounded by the complexity and heterogeneity of nomenclatures used to describe diseases, genes and genetic variants. Mastermind is a widely-used bioinformatic platform of genomic associations that has indexed more than 7.5 M full-text articles and 2.5 M supplemental datasets. It has automatically identified, disambiguated and annotated >6.1 M genetic variants and identified >50 K disease-gene associations. Here, we describe how Mastermind improves the sensitivity and reproducibility of clinical variant interpretation and produces comprehensive genomic landscapes of genetic variants driving pharmaceutical research. We demonstrate an alarmingly high degree of heterogeneity across commercially available panels for hereditary cancer that is resolved by evidence from Mastermind. We further examined the sensitivity of Mastermind for variant interpretation by examining 108 clinically-encountered variants and comparing the results to alternate methods. Mastermind demonstrated a sensitivity of 98.4% compared to 4.4, 45.6, and 37.4% for alternatives PubMed, Google Scholar, and ClinVar, respectively, and a specificity of 98.5% compared to 45.1, 57.6, and 68.8% as well as an increase in content yield of 22.6-, 2.2-, and 2.6-fold. When curated for clinical significance, Mastermind identified more than 4.9-fold more pathogenic variants than ClinVar for representative genes. For structural variants, we compared Mastermind's ability to sensitively identify evidence for 10 representative disease-causing CNVs versus results identified in PubMed, as well as its ability to identify evidence for fusion events compared to COSMIC. Mastermind demonstrated a 4.0- to 43.9-fold increase in references for specific CNVs compared to PubMed, as well as 5.4-fold more fusion genes when compared with COSMIC's curated database. Additionally, Mastermind produced an 8.0-fold increase in reference citations for fusion events common to Mastermind and outside databases. Taken together, these results demonstrate the utility and superiority of Mastermind in terms of both sensitivity and specificity of automated results for clinical diagnostic variant interpretation for multiple genetic variant types and highlight the potential benefit in informing pharmaceutical research.

8.
J Low Genit Tract Dis ; 23(3): 214-219, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31232912

RESUMO

OBJECTIVE: Vulvar lichen sclerosus (LS) is known to occur in families, suggesting a genetic link. Genomic profiling of patients with vulvar LS was investigated to find underlying pathogenetic mechanisms, with the hope that targeted therapies and future clinical research will arise. METHODS: Two unrelated families with vulvar LS were investigated using whole-exome sequencing. Five affected sisters from 1 family were compared with their unaffected paternal aunt (unaffected control). A mother-daughter pair from a second affected family was compared with the first family. The results of the sequencing were compared with population-specific allele frequency databases to prioritize potential variants contributing to vulvar LS development. RESULTS: Recurrent germ-line variants in 4 genes were identified as likely to be deleterious to proper protein function in all of the 7 affected patients, but not in the unaffected control. The genes with variants included CD177 (neutrophil activation), CD200 (inhibitory signal to macrophages), ANKRD18A (ankyrin repeat protein, epigenetic regulation), and LATS2 (co-repressor of androgen signaling). CONCLUSIONS: Although many providers may see a mother and daughter with vulvar LS, this condition is rarely seen in multiple family members who are available for genetic testing. This is the first report to detail genomic profiling related to a familial association of vulvar LS.


Assuntos
Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Líquen Escleroso Vulvar/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Frequência do Gene , Humanos , Masculino , Pessoa de Meia-Idade
9.
Gastroenterology ; 154(4): 897-905.e1, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29146522

RESUMO

BACKGROUND & AIMS: The incidence of colorectal cancer (CRC) in individuals younger than 50 years is increasing. We sought to ascertain the proportion of young CRC cases associated with genetic predisposition. METHODS: We performed a retrospective study of individuals diagnosed with CRC at an age younger than 50 years, evaluated by the clinical genetics service at a single tertiary care cancer center from 1998 through 2015. We collected data on patient histories, tumor phenotypes, and results of germline DNA sequencing. For subjects with uninformative clinical evaluations, germline DNA samples were (re)sequenced using a research-based next-generation sequencing multigene panel. The primary outcome was identification of a pathogenic germline mutation associated with cancer predisposition. RESULTS: Of 430 young CRC cases, 111 (26%) had a first-degree relative with CRC. Forty-one of the subjects with CRC (10%) had tumors with histologic evidence for mismatch repair deficiency. Of 315 subjects who underwent clinical germline sequencing, 79 had mutations associated with a hereditary cancer syndrome and 21 had variants of uncertain significance. Fifty-six subjects had pathogenic variants associated with Lynch syndrome (25 with mutations in MSH2, 24 with mutations in MLH1, 5 with mutations in MSH6, and 2 with mutations in PMS2) and 10 subjects had pathogenic variants associated with familial adenomatous polyposis. Thirteen subjects had mutations in other cancer-associated genes (8 in MUTYH, 2 in SMAD4, 1 in BRCA1, 1 in TP53, and 1 in CHEK2), all identified through multigene panel tests. Among 117 patients with uninformative clinical evaluations, next-generation sequence analysis using a multigene panel detected actionable germline variants in 6 patients (5%). Only 43 of the 85 subjects with germline mutations associated with a hereditary cancer syndrome (51%) reported a CRC diagnosis in a first-degree relative. CONCLUSIONS: Approximately 1 in 5 individuals diagnosed with CRC at age younger than 50 years carries a germline mutation associated with cancer; nearly half of these do not have clinical histories typically associated with the identified syndrome. Germline testing with multigene cancer panels should be considered for all young patients with CRC.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias Colorretais/genética , Mutação em Linhagem Germinativa , Adolescente , Adulto , Idade de Início , Neoplasias Colorretais/diagnóstico , Análise Mutacional de DNA/métodos , Feminino , Predisposição Genética para Doença , Hereditariedade , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Pessoa de Meia-Idade , Linhagem , Fenótipo , Valor Preditivo dos Testes , Estudos Retrospectivos , Fatores de Risco , Centros de Atenção Terciária , Adulto Jovem
10.
Nat Commun ; 6: 8470, 2015 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-26415585

RESUMO

Sézary syndrome (SS) is an aggressive leukaemia of mature T cells with poor prognosis and limited options for targeted therapies. The comprehensive genetic alterations underlying the pathogenesis of SS are unknown. Here we integrate whole-genome sequencing (n=6), whole-exome sequencing (n=66) and array comparative genomic hybridization-based copy-number analysis (n=80) of primary SS samples. We identify previously unknown recurrent loss-of-function aberrations targeting members of the chromatin remodelling/histone modification and trithorax families, including ARID1A in which functional loss from nonsense and frameshift mutations and/or targeted deletions is observed in 40.3% of SS genomes. We also identify recurrent gain-of-function mutations targeting PLCG1 (9%) and JAK1, JAK3, STAT3 and STAT5B (JAK/STAT total ∼11%). Functional studies reveal sensitivity of JAK1-mutated primary SS cells to JAK inhibitor treatment. These results highlight the complex genomic landscape of SS and a role for inhibition of JAK/STAT pathways for the treatment of SS.


Assuntos
Epigênese Genética/genética , Janus Quinases/genética , Fatores de Transcrição STAT/genética , Síndrome de Sézary/genética , Proteínas Adaptadoras de Sinalização CARD/genética , Proteínas de Ciclo Celular/genética , Variações do Número de Cópias de DNA , Análise Mutacional de DNA , Proteínas de Ligação a DNA/genética , Exoma , Genômica , Guanilato Ciclase/genética , Humanos , Histona Desmetilases com o Domínio Jumonji/genética , Células Jurkat , Família Multigênica , Proteínas de Neoplasias/genética , Fosfolipase C gama/genética , Proteínas ras/genética
11.
Clin Infect Dis ; 61(2): 233-41, 2015 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-25828993

RESUMO

BACKGROUND: Clostridium difficile infection (CDI) can cause severe disease and death, especially in older adults. A better understanding of risk factors for adverse outcomes is needed. This study tests the hypotheses that infection with specific ribotypes and presence of stool toxins independently associate with severity and constructs predictive models of adverse outcomes. METHODS: Cases of non-recurrent CDI were prospectively included after positive stool tests for toxins A and/or B by enzyme immunoassay (EIA) or tcdB by polymerase chain reaction. Outcomes included severe CDI (intensive care unit admission, colectomy, or death attributable to CDI within 30 days of diagnosis) and 30-day all-cause mortality. Adjusted models were developed to test hypotheses and predict outcomes. RESULTS: In total, 1144 cases were included. The toxin EIA was positive in 37.2% and 35.6% of patients were of age >65 years. One of the 137 unique ribotypes was ribotype 027 (16.2%). Detectable stool toxin did not associate with outcomes. Adjusting for covariates, including age, Ribotype 027 was a significant predictor of severe CDI (90 cases; odds ratio [OR], 1.73; 95% confidence interval [CI], 1.03-2.89; P = .037) and mortality (89 cases; OR, 2.02; 95% CI, 1.19-3.43; P = .009). Concurrent antibiotic use associated with both outcomes. Both multivariable predictive models had excellent performance (area under the curve >0.8). CONCLUSIONS: Detection of stool toxin A and/or B by EIA does not predict severe CDI or mortality. Infection with ribotype 027 independently predicts severe CDI and mortality. Use of concurrent antibiotics is a potentially modifiable risk factor for severe CDI.


Assuntos
Toxinas Botulínicas/isolamento & purificação , Clostridioides difficile/genética , Clostridioides difficile/patogenicidade , Enterocolite Pseudomembranosa/diagnóstico , Enterocolite Pseudomembranosa/mortalidade , Fezes/microbiologia , Ribotipagem , Adulto , Fatores Etários , Idoso , Clostridioides difficile/classificação , Enterocolite Pseudomembranosa/microbiologia , Feminino , Humanos , Técnicas Imunoenzimáticas , Masculino , Pessoa de Meia-Idade , Modelos Estatísticos , Razão de Chances , Reação em Cadeia da Polimerase , Curva ROC , Fatores de Risco , Índice de Gravidade de Doença
12.
Blood ; 124(25): 3768-71, 2014 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-25349176

RESUMO

The spectrum of cutaneous CD30-positive lymphoproliferative disorders (LPDs) includes lymphomatoid papulosis and primary cutaneous anaplastic large cell lymphoma. Chromosomal translocations targeting tyrosine kinases in CD30-positive LPDs have not been described. Using whole-transcriptome sequencing, we identified a chimeric fusion involving NPM1 (5q35) and TYK2 (19p13) that encodes an NPM1-TYK2 protein containing the oligomerization domain of NPM1 and an intact catalytic domain in TYK2. Fluorescence in situ hybridization revealed NPM1-TYK2 fusions in 2 of 47 (4%) primary cases of CD30-positive LPDs and was absent in other mature T-cell neoplasms (n = 151). Functionally, NPM1-TYK2 induced constitutive TYK2, signal transducer and activator of transcription 1 (STAT1), STAT3, and STAT5 activation. Conversely, a kinase-defective NPM1-TYK2 mutant abrogated STAT1/3/5 signaling. Finally, short hairpin RNA-mediated silencing of TYK2 abrogated lymphoma cell growth. This is the first report of recurrent translocations involving TYK2, and it highlights the novel therapeutic opportunities in the treatment of CD30-positive LPDs with TYK2 translocations.


Assuntos
Antígeno Ki-1/genética , Linfoma Anaplásico Cutâneo Primário de Células Grandes/genética , Papulose Linfomatoide/genética , Proteínas Nucleares/genética , Proteínas de Fusão Oncogênica/genética , Neoplasias Cutâneas/genética , TYK2 Quinase/genética , Western Blotting , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Hibridização in Situ Fluorescente , Antígeno Ki-1/metabolismo , Linfoma Anaplásico Cutâneo Primário de Células Grandes/metabolismo , Linfoma Anaplásico Cutâneo Primário de Células Grandes/patologia , Papulose Linfomatoide/metabolismo , Papulose Linfomatoide/patologia , Mutação , Proteínas Nucleares/metabolismo , Nucleofosmina , Fusão Oncogênica , Proteínas de Fusão Oncogênica/metabolismo , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais/genética , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , TYK2 Quinase/metabolismo , Transcriptoma/genética
13.
Blood ; 124(10): 1655-8, 2014 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-24982505

RESUMO

Langerhans cell histiocytosis (LCH) represents a clonal proliferation of Langerhans cells. BRAF V600E mutations have been identified in approximately 50% of cases. To discover other genetic mechanisms underlying LCH pathogenesis, we studied 8 cases of LCH using a targeted next-generation sequencing platform. An E102_I103del mutation in MAP2K1 was identified in one BRAF wild-type case and confirmed by Sanger sequencing. Analysis of 32 additional cases using BRAF V600E allele-specific polymerase chain reaction and Sanger sequencing of MAP2K1 exons 2 and 3 revealed somatic, mutually exclusive BRAF and MAP2K1 mutations in 18 of 40 (45.0%) and 11 of 40 (27.5%) cases, respectively. This is the first report of MAP2K1 mutations in LCH that occur in 50% of BRAF wild-type cases. The mutually exclusive nature of MAP2K1 and BRAF mutations implicates a critical role of oncogenic MAPK signaling in LCH. This finding may also have implications in the use of BRAF and MEK inhibitor therapy.


Assuntos
Histiocitose de Células de Langerhans/genética , MAP Quinase Quinase 1/genética , Proteínas Proto-Oncogênicas B-raf/genética , Substituição de Aminoácidos , Feminino , Frequência do Gene , Predisposição Genética para Doença , Ácido Glutâmico/genética , Histiocitose de Células de Langerhans/epidemiologia , Humanos , Masculino , Mutação de Sentido Incorreto , Prevalência , Estudos Retrospectivos , Valina/genética
14.
Blood ; 124(9): 1460-72, 2014 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-24825865

RESUMO

The comprehensive genetic alterations underlying the pathogenesis of T-cell prolymphocytic leukemia (T-PLL) are unknown. To address this, we performed whole-genome sequencing (WGS), whole-exome sequencing (WES), high-resolution copy-number analysis, and Sanger resequencing of a large cohort of T-PLL. WGS and WES identified novel mutations in recurrently altered genes not previously implicated in T-PLL including EZH2, FBXW10, and CHEK2. Strikingly, WGS and/or WES showed largely mutually exclusive mutations affecting IL2RG, JAK1, JAK3, or STAT5B in 38 of 50 T-PLL genomes (76.0%). Notably, gain-of-function IL2RG mutations are novel and have not been reported in any form of cancer. Further, high-frequency mutations in STAT5B have not been previously reported in T-PLL. Functionally, IL2RG-JAK1-JAK3-STAT5B mutations led to signal transducer and activator of transcription 5 (STAT5) hyperactivation, transformed Ba/F3 cells resulting in cytokine-independent growth, and/or enhanced colony formation in Jurkat T cells. Importantly, primary T-PLL cells exhibited constitutive activation of STAT5, and targeted pharmacologic inhibition of STAT5 with pimozide induced apoptosis in primary T-PLL cells. These results for the first time provide a portrait of the mutational landscape of T-PLL and implicate deregulation of DNA repair and epigenetic modulators as well as high-frequency mutational activation of the IL2RG-JAK1-JAK3-STAT5B axis in the pathogenesis of T-PLL. These findings offer opportunities for novel targeted therapies in this aggressive leukemia.


Assuntos
Leucemia Prolinfocítica de Células T/genética , Mutação , Adulto , Idoso , Idoso de 80 Anos ou mais , Sequência de Aminoácidos , Proteínas Mutadas de Ataxia Telangiectasia/genética , Sequência de Bases , Morte Celular/efeitos dos fármacos , Estudos de Coortes , Simulação por Computador , Variações do Número de Cópias de DNA , Análise Mutacional de DNA , DNA de Neoplasias/genética , Exoma , Feminino , Humanos , Subunidade gama Comum de Receptores de Interleucina/genética , Janus Quinase 1/genética , Janus Quinase 3/química , Janus Quinase 3/genética , Leucemia Prolinfocítica de Células T/tratamento farmacológico , Leucemia Prolinfocítica de Células T/metabolismo , Masculino , Pessoa de Meia-Idade , Modelos Moleculares , Dados de Sequência Molecular , Pimozida/farmacologia , Conformação Proteica , Fator de Transcrição STAT5/antagonistas & inibidores , Fator de Transcrição STAT5/química , Fator de Transcrição STAT5/genética , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Células Tumorais Cultivadas
15.
J Exp Med ; 209(9): 1553-65, 2012 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-22891276

RESUMO

Splenic marginal zone lymphoma (SMZL), the most common primary lymphoma of spleen, is poorly understood at the genetic level. In this study, using whole-genome DNA sequencing (WGS) and confirmation by Sanger sequencing, we observed mutations identified in several genes not previously known to be recurrently altered in SMZL. In particular, we identified recurrent somatic gain-of-function mutations in NOTCH2, a gene encoding a protein required for marginal zone B cell development, in 25 of 99 (∼25%) cases of SMZL and in 1 of 19 (∼5%) cases of nonsplenic MZLs. These mutations clustered near the C-terminal proline/glutamate/serine/threonine (PEST)-rich domain, resulting in protein truncation or, rarely, were nonsynonymous substitutions affecting the extracellular heterodimerization domain (HD). NOTCH2 mutations were not present in other B cell lymphomas and leukemias, such as chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL; n = 15), mantle cell lymphoma (MCL; n = 15), low-grade follicular lymphoma (FL; n = 44), hairy cell leukemia (HCL; n = 15), and reactive lymphoid hyperplasia (n = 14). NOTCH2 mutations were associated with adverse clinical outcomes (relapse, histological transformation, and/or death) among SMZL patients (P = 0.002). These results suggest that NOTCH2 mutations play a role in the pathogenesis and progression of SMZL and are associated with a poor prognosis.


Assuntos
Linfoma de Células B/genética , Mutação , Receptor Notch2/genética , Neoplasias Esplênicas/genética , Idoso , Feminino , Genoma Humano , Humanos , Linfoma de Células B/diagnóstico , Linfoma de Células B/mortalidade , Masculino , Pessoa de Meia-Idade , Estrutura Terciária de Proteína , Receptor Notch2/metabolismo , Análise de Sequência de DNA , Neoplasias Esplênicas/diagnóstico , Neoplasias Esplênicas/mortalidade , Taxa de Sobrevida
16.
Arch Pathol Lab Med ; 136(8): 871-5, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22849733

RESUMO

We report a case of indolent mantle cell lymphoma with progression to pleomorphic mantle cell lymphoma 8 years after initial presentation. The first lymph node biopsy showed expanded mantle zones composed of uniformly small B lymphocytes. A cyclin D1 immunohistochemical stain was negative and the patient was observed. Eight years later, the patient developed symptomatic splenomegaly. Microscopic examination of the spleen revealed expanded mantle zones with an increased number of large cells with irregular nuclear contours. Immunohistochemistry for cyclin D1 was positive. A repeat cyclin D1 immunohistochemical staining performed on the initial lymph node biopsy was positive, indicating an inadequate initial study. Immunoglobulin heavy-chain gene rearrangement studies confirmed clonal identity. A revised diagnosis of indolent mantle cell lymphoma with progression to pleomorphic mantle cell lymphoma was rendered. The differential diagnosis of mantle cell lymphoma, including clinical and morphologic variants, is discussed.


Assuntos
Transformação Celular Neoplásica , Linfonodos/patologia , Linfoma de Célula do Manto/diagnóstico , Linfoma de Célula do Manto/patologia , Ciclina D1/metabolismo , Diagnóstico Diferencial , Rearranjo Gênico , Genes de Cadeia Pesada de Imunoglobulina , Humanos , Linfonodos/metabolismo , Linfoma de Célula do Manto/genética , Linfoma de Célula do Manto/metabolismo , Masculino , Pessoa de Meia-Idade , Proteínas de Neoplasias/metabolismo
17.
Blood ; 115(13): 2592-600, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20110425

RESUMO

Because the microenvironment that supports hematopoietic stem cell (HSC) proliferation and differentiation is not fully understood, we adapted a heterotopic bone formation model as a new approach for studying the HSC microenvironment in vivo. Endogenous HSCs homed to tissue-engineered ossicles and individually sorted HSCs from ossicles were able to reconstitute lethally irradiated mice. To further explore this model as a system to study the stem cell niche, ossicles were established with or without anabolic parathyroid hormone (PTH) treatment during the 4-week course of bone development. Histology and micro-computed tomography showed higher bone area-to-total area ratios, thicker cortical bone and trabecular bone, significantly higher bone mineral density and bone volume fraction in PTH-treated groups than in controls. By an in vivo competitive long-term reconstitution assay, HSC frequency in the ossicle marrow was 3 times greater in PTH groups than in controls. When whole bone marrow cells were directly injected into the ossicles after lethal irradiation, the PTH-treated groups showed an enhanced reconstitution rate compared with controls. These findings suggest the residence of HSCs in heterotopic bone marrow and support the future use of this ossicle model in elucidating the composition and regulation of the HSC niche.


Assuntos
Células-Tronco Hematopoéticas/citologia , Modelos Animais , Ossificação Heterotópica , Adipócitos/fisiologia , Animais , Biomarcadores , Medula Óssea/ultraestrutura , Contagem de Células , Linhagem da Célula , Células Cultivadas/transplante , Transplante de Células-Tronco Hematopoéticas/métodos , Megacariócitos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Hormônio Paratireóideo/farmacologia , Quimera por Radiação , Células Estromais/fisiologia , Tela Subcutânea , Alicerces Teciduais
18.
Cell Stem Cell ; 4(2): 170-9, 2009 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-19119091

RESUMO

According to the "osteoblastic niche" model, hematopoietic stem cells (HSCs) are maintained by N-cadherin-mediated homophilic adhesion to osteoblasts at the bone marrow endosteum. In contrast to this model, we cannot detect N-cadherin expression by HSCs, and most HSCs do not localize to the endosteal surface. It has nonetheless been suggested that HSCs express low levels of N-cadherin that regulate HSC maintenance. To test this, we conditionally deleted N-cadherin from HSCs and other hematopoietic cells in adult Mx-1-Cre(+)N-cadherin(fl/-) mice. N-cadherin deficiency had no detectable effect on HSC maintenance or hematopoiesis. N-cadherin deficiency did not affect bone marrow cellularity or lineage composition, the numbers of colony-forming progenitors, the frequency of HSCs, the ability of HSCs to sustain hematopoiesis over time, or their ability to reconstitute irradiated mice in primary or secondary transplants. Loss of N-cadherin does not lead to HSC depletion. N-cadherin expression by HSCs is not necessary for niche function.


Assuntos
Caderinas/metabolismo , Células-Tronco Hematopoéticas/fisiologia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/fisiologia , Caderinas/genética , Linhagem da Célula , Células Cultivadas , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Radiação Ionizante , Transplante de Células-Tronco
20.
Nat Rev Immunol ; 8(4): 290-301, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18323850

RESUMO

Haematopoietic stem cell (HSC) niches are specialized microenvironments that contain stem cells and regulate their maintenance. Cells at the interface of bone and the bone marrow (the endosteum) contribute to the creation of HSC niches. It remains uncertain whether this interface itself is a niche, or whether endosteal cells secrete factors that diffuse to nearby niches. Vascular and/or perivascular cells may also create niches as many HSCs are observed around sinusoidal blood vessels, and perivascular cells secrete factors that regulate HSC maintenance. Do endosteal and perivascular cells create distinct niches, or do they contribute to a common niche? We discuss a range of niche models consistent with recent evidence.


Assuntos
Células da Medula Óssea/citologia , Hematopoese , Células-Tronco Hematopoéticas/citologia , Animais , Vasos Sanguíneos/citologia , Medula Óssea/anatomia & histologia , Osso e Ossos/citologia , Diferenciação Celular/fisiologia , Humanos , Osteoblastos/citologia , Osteoclastos/citologia , Baço/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...