Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Am J Hum Genet ; 108(9): 1578-1589, 2021 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-34265237

RESUMO

Thoracic aortic aneurysm (TAA) is characterized by dilation of the aortic root or ascending/descending aorta. TAA is a heritable disease that can be potentially life threatening. While 10%-20% of TAA cases are caused by rare, pathogenic variants in single genes, the origin of the majority of TAA cases remains unknown. A previous study implicated common variants in FBN1 with TAA disease risk. Here, we report a genome-wide scan of 1,351 TAA-affected individuals and 18,295 control individuals from the Cardiovascular Health Improvement Project and Michigan Genomics Initiative at the University of Michigan. We identified a genome-wide significant association with TAA for variants within the third intron of TCF7L2 following replication with meta-analysis of four additional independent cohorts. Common variants in this locus are the strongest known genetic risk factor for type 2 diabetes. Although evidence indicates the presence of different causal variants for TAA and type 2 diabetes at this locus, we observed an opposite direction of effect. The genetic association for TAA colocalizes with an aortic eQTL of TCF7L2, suggesting a functional relationship. These analyses predict an association of higher expression of TCF7L2 with TAA disease risk. In vitro, we show that upregulation of TCF7L2 is associated with BCL2 repression promoting vascular smooth muscle cell apoptosis, a key driver of TAA disease.


Assuntos
Aneurisma da Aorta Torácica/genética , Diabetes Mellitus Tipo 2/genética , Células Endoteliais/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Locos de Características Quantitativas , Proteína 2 Semelhante ao Fator 7 de Transcrição/genética , Aorta/metabolismo , Aorta/patologia , Aneurisma da Aorta Torácica/metabolismo , Aneurisma da Aorta Torácica/patologia , Estudos de Casos e Controles , Caspase 3/genética , Caspase 3/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Células Endoteliais/patologia , Regulação da Expressão Gênica , Genoma Humano , Estudo de Associação Genômica Ampla , Humanos , Íntrons , Michigan , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Mutação , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína 2 Semelhante ao Fator 7 de Transcrição/metabolismo , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
2.
Blood ; 128(15): 1913-1917, 2016 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-27488349

RESUMO

The congenital sideroblastic anemias (CSAs) are a heterogeneous group of inherited blood disorders characterized by pathological mitochondrial iron deposition in erythroid precursors. Each known cause has been attributed to a mutation in a protein associated with heme biosynthesis, iron-sulfur cluster biogenesis, mitochondrial translation, or a component of the mitochondrial respiratory chain. Here, we describe a recurring mutation, c.276_278del, p.F93del, in NDUFB11, a mitochondrial respiratory complex I-associated protein encoded on the X chromosome, in 5 males with a variably syndromic, normocytic CSA. The p.F93del mutation results in respiratory insufficiency and loss of complex I stability and activity in patient-derived fibroblasts. Targeted introduction of this allele into K562 erythroleukemia cells results in a proliferation defect with minimal effect on erythroid differentiation potential, suggesting the mechanism of anemia in this disorder.


Assuntos
Anemia Sideroblástica/genética , Sequência de Bases , Cromossomos Humanos X/genética , Complexo I de Transporte de Elétrons/genética , Doenças Genéticas Ligadas ao Cromossomo X/genética , Deleção de Sequência , Adolescente , Adulto , Idoso , Anemia Sideroblástica/metabolismo , Anemia Sideroblástica/patologia , Criança , Pré-Escolar , Cromossomos Humanos X/metabolismo , Complexo I de Transporte de Elétrons/metabolismo , Feminino , Doenças Genéticas Ligadas ao Cromossomo X/metabolismo , Humanos , Células K562 , Masculino , Pessoa de Meia-Idade
3.
Blood ; 126(25): 2734-8, 2015 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-26491070

RESUMO

The congenital sideroblastic anemias (CSAs) are relatively uncommon diseases characterized by defects in mitochondrial heme synthesis, iron-sulfur (Fe-S) cluster biogenesis, or protein synthesis. Here we demonstrate that mutations in HSPA9, a mitochondrial HSP70 homolog located in the chromosome 5q deletion syndrome 5q33 critical deletion interval and involved in mitochondrial Fe-S biogenesis, result in CSA inherited as an autosomal recessive trait. In a fraction of patients with just 1 severe loss-of-function allele, expression of the clinical phenotype is associated with a common coding single nucleotide polymorphism in trans that correlates with reduced messenger RNA expression and results in a pseudodominant pattern of inheritance.


Assuntos
Anemia Sideroblástica/genética , Doenças Genéticas Ligadas ao Cromossomo X/genética , Proteínas de Choque Térmico HSP70/genética , Proteínas Mitocondriais/genética , Adulto , Idoso , Sequência de Bases , Análise Mutacional de DNA , Feminino , Genótipo , Humanos , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Mutação , Análise de Sequência com Séries de Oligonucleotídeos , Linhagem , Polimorfismo de Nucleotídeo Único , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Adulto Jovem
5.
Blood ; 124(18): 2867-71, 2014 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-25193871

RESUMO

Mutations in genes encoding proteins that are involved in mitochondrial heme synthesis, iron-sulfur cluster biogenesis, and mitochondrial protein synthesis have previously been implicated in the pathogenesis of the congenital sideroblastic anemias (CSAs). We recently described a syndromic form of CSA associated with B-cell immunodeficiency, periodic fevers, and developmental delay (SIFD). Here we demonstrate that SIFD is caused by biallelic mutations in TRNT1, the gene encoding the CCA-adding enzyme essential for maturation of both nuclear and mitochondrial transfer RNAs. Using budding yeast lacking the TRNT1 homolog, CCA1, we confirm that the patient-associated TRNT1 mutations result in partial loss of function of TRNT1 and lead to metabolic defects in both the mitochondria and cytosol, which can account for the phenotypic pleiotropy.


Assuntos
Anemia Sideroblástica/congênito , Anemia Sideroblástica/genética , Deficiências do Desenvolvimento/complicações , Febre/complicações , Doenças Genéticas Ligadas ao Cromossomo X/genética , Síndromes de Imunodeficiência/complicações , Mutação/genética , RNA Nucleotidiltransferases/genética , Alelos , Anemia Sideroblástica/complicações , Anemia Sideroblástica/enzimologia , Deficiências do Desenvolvimento/genética , Febre/genética , Doenças Genéticas Ligadas ao Cromossomo X/complicações , Doenças Genéticas Ligadas ao Cromossomo X/enzimologia , Células HEK293 , Humanos , Síndromes de Imunodeficiência/genética
6.
Proc Natl Acad Sci U S A ; 110(14): 5504-9, 2013 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-23520050

RESUMO

Sulfolobus turreted icosahedral virus (STIV) was isolated in acidic hot springs where it infects the archeon Sulfolobus solfataricus. We determined the STIV structure using near-atomic resolution electron microscopy and X-ray crystallography allowing tracing of structural polypeptide chains and visualization of transmembrane proteins embedded in the viral membrane. We propose that the vertex complexes orchestrate virion assembly by coordinating interactions of the membrane and various protein components involved. STIV shares the same coat subunit and penton base protein folds as some eukaryotic and bacterial viruses, suggesting that they derive from a common ancestor predating the divergence of the three kingdoms of life. One architectural motif (ß-jelly roll fold) forms virtually the entire capsid (distributed in three different gene products), indicating that a single ancestral protein module may have been at the origin of its evolution.


Assuntos
Proteínas de Membrana/química , Modelos Moleculares , Rudiviridae/química , Sulfolobus/virologia , Microscopia Crioeletrônica , Cristalografia por Raios X
7.
Blood ; 121(7): 1200-8, 2013 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-23223430

RESUMO

Mutations in HFE lead to hereditary hemochromatosis (HH) because of inappropriately high iron uptake from the diet resulting from decreased hepatic expression of the iron-regulatory hormone hepcidin. -thalassemia is a congenital anemia caused by partial or complete loss of -globin synthesis causing ineffective erythropoiesis, anemia, decreased hepcidin production, and secondary iron overload. Tmprss6 is postulated to regulate hepcidin production by cleaving Hemojuvelin (Hjv), a key modulator of hepcidin expression, from the hepatocyte surface. On this basis, we hypothesized that treatment of mouse models of HH (Hfe(-/-)) and -thalassemia intermedia (Hbb(th3/+)) with Tmprss6 siRNA formulated in lipid nanoparticles (LNPs) that are preferentially taken up by the liver would increase hepcidin expression and lessen the iron loading in both models. In the present study, we demonstrate that LNP-Tmprss6 siRNA treatment of Hfe(-/-) and Hbb(th3/+) mice induces hepcidin and diminishes tissue and serum iron levels. Furthermore, LNP-Tmprss6 siRNA treatment of Hbb(th3/+) mice substantially improved the anemia by altering RBC survival and ineffective erythropoiesis. Our results indicate that pharmacologic manipulation of Tmprss6 with RNAi therapeutics isa practical approach to treating iron overload diseases associated with diminished hepcidin expression and may have efficacy in modifying disease-associated morbidities of -thalassemia intermedia.


Assuntos
Sobrecarga de Ferro/terapia , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/deficiência , Talassemia beta/terapia , Anemia/genética , Anemia/metabolismo , Anemia/terapia , Animais , Peptídeos Catiônicos Antimicrobianos/genética , Peptídeos Catiônicos Antimicrobianos/metabolismo , Sequência de Bases , Modelos Animais de Doenças , Envelhecimento Eritrocítico , Eritropoese , Feminino , Hemocromatose/genética , Hemocromatose/metabolismo , Hemocromatose/terapia , Proteína da Hemocromatose , Hepcidinas , Antígenos de Histocompatibilidade Classe I/genética , Sobrecarga de Ferro/genética , Sobrecarga de Ferro/metabolismo , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nanopartículas , Interferência de RNA , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/uso terapêutico , Serina Endopeptidases/genética , Talassemia beta/genética , Talassemia beta/metabolismo
8.
J Biol Chem ; 288(28): 20668-82, 2013 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-23733181

RESUMO

Steap4 is a cell surface metalloreductase linked to obesity-associated insulin resistance. Initial characterization of its cell surface metalloreductase activity has been reported, but thorough biochemical characterization of this activity is lacking. Here, we report detailed kinetic analysis of the Steap4 cell surface metalloreductase activities. Steap4 shows physiologically relevant Km values for both Fe(3+) and Cu(2+) and retains activity at acidic pH, suggesting it may also function within intracellular organelles to reduce these metals. Flavin-dependent NADPH oxidase activity that was much greater than the equivalent Steap3 construct was observed for the isolated N-terminal oxidoreductase domain. The crystal structure of the Steap4 oxidoreductase domain was determined, providing a structural explanation for these differing activities. Structure-function work also suggested Steap4 utilizes an interdomain flavin-binding site to shuttle electrons between the oxidoreductase and transmembrane domains, and it showed that the disordered N-terminal residues do not contribute to enzymatic activity.


Assuntos
Cobre/metabolismo , Flavinas/metabolismo , Ferro/metabolismo , Proteínas de Membrana/metabolismo , Oxirredutases/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação/genética , Cristalografia por Raios X , Eletroforese em Gel de Poliacrilamida , Endossomos/enzimologia , Endossomos/metabolismo , Células HEK293 , Humanos , Concentração de Íons de Hidrogênio , Cinética , Lisossomos/enzimologia , Lisossomos/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/genética , Microscopia de Fluorescência , Modelos Moleculares , Dados de Sequência Molecular , NADPH Oxidases/genética , NADPH Oxidases/metabolismo , Oxirredutases/química , Oxirredutases/genética , Estrutura Terciária de Proteína , Ratos , Homologia de Sequência de Aminoácidos
9.
Am J Hematol ; 89(3): 315-9, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24166784

RESUMO

X-linked sideroblastic anemia (XLSA) is the most common form of congenital sideroblastic anemia. In affected males, it is uniformly associated with partial loss-of-function missense mutations in the erythroid-specific heme biosynthesis protein 5-aminolevulinate synthase 2 (ALAS2). Here, we report five families with XLSA owing to mutations in a GATA transcription factor binding site located in a transcriptional enhancer element in intron 1 of the ALAS2 gene. As such, this study defines a new class of mutations that should be evaluated in patients undergoing genetic testing for a suspected diagnosis of XLSA.


Assuntos
5-Aminolevulinato Sintetase/genética , Anemia Sideroblástica/genética , Elementos Facilitadores Genéticos/genética , Fatores de Transcrição GATA/metabolismo , Doenças Genéticas Ligadas ao Cromossomo X/genética , Íntrons/genética , Mutação , Adulto , Idoso , Anemia Sideroblástica/sangue , Sítios de Ligação , Europa (Continente)/etnologia , Feminino , Doenças Genéticas Ligadas ao Cromossomo X/sangue , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Linhagem , Adulto Jovem
10.
Nat Genet ; 55(7): 1106-1115, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37308786

RESUMO

The current understanding of the genetic determinants of thoracic aortic aneurysms and dissections (TAAD) has largely been informed through studies of rare, Mendelian forms of disease. Here, we conducted a genome-wide association study (GWAS) of TAAD, testing ~25 million DNA sequence variants in 8,626 participants with and 453,043 participants without TAAD in the Million Veteran Program, with replication in an independent sample of 4,459 individuals with and 512,463 without TAAD from six cohorts. We identified 21 TAAD risk loci, 17 of which have not been previously reported. We leverage multiple downstream analytic methods to identify causal TAAD risk genes and cell types and provide human genetic evidence that TAAD is a non-atherosclerotic aortic disorder distinct from other forms of vascular disease. Our results demonstrate that the genetic architecture of TAAD mirrors that of other complex traits and that it is not solely inherited through protein-altering variants of large effect size.


Assuntos
Aneurisma da Aorta Torácica , Dissecção Aórtica , Veteranos , Humanos , Estudo de Associação Genômica Ampla , Linhagem , Aneurisma da Aorta Torácica/genética , Dissecção Aórtica/genética
11.
Proc Natl Acad Sci U S A ; 105(21): 7410-5, 2008 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-18495927

RESUMO

The daily production of 200 billion erythrocytes requires 20 mg of iron, accounting for nearly 80% of the iron demand in humans. Thus, erythroid precursor cells possess an efficient mechanism for iron uptake in which iron loaded transferrin (Tf) binds to the transferrin receptor (TfR) at the cell surface. The Tf:TfR complex then enters the endosome via receptor-mediated endocytosis. Upon endosomal acidification, iron is released from Tf, reduced to Fe(2+) by Steap3, and transported across the endosomal membrane by divalent metal iron transporter 1. Steap3, the major ferrireductase in erythrocyte endosomes, is a member of a unique family of reductases. Steap3 is comprised of an N-terminal cytosolic oxidoreductase domain and a C-terminal heme-containing transmembrane domain. Cytosolic NADPH and a flavin are predicted cofactors, but the NADPH/flavin binding domain differs significantly from those in other eukaryotic reductases. Instead, Steap3 shows remarkable, although limited homology to FNO, an archaeal oxidoreductase. We have determined the crystal structure of the human Steap3 oxidoreductase domain in the absence and presence of NADPH. The structure reveals an FNO-like domain with an unexpected dimer interface and substrate binding sites that are well positioned to direct electron transfer from the cytosol to a heme moiety predicted to be fixed within the transmembrane domain. Here, we discuss possible gating mechanisms for electron transfer across the endosomal membrane.


Assuntos
FMN Redutase/química , Proteínas Oncogênicas/química , Oxirredutases/química , Sítios de Ligação , Proteínas de Ciclo Celular , Cristalografia , Dimerização , Células Precursoras Eritroides/enzimologia , Humanos , NADP/química , Estrutura Terciária de Proteína , Eletricidade Estática , Transferrina/química
12.
J Clin Invest ; 130(10): 5245-5256, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32634119

RESUMO

The congenital sideroblastic anemias (CSAs) can be caused by primary defects in mitochondrial iron-sulfur (Fe-S) cluster biogenesis. HSCB (heat shock cognate B), which encodes a mitochondrial cochaperone, also known as HSC20 (heat shock cognate protein 20), is the partner of mitochondrial heat shock protein A9 (HSPA9). Together with glutaredoxin 5 (GLRX5), HSCB and HSPA9 facilitate the transfer of nascent 2-iron, 2-sulfur clusters to recipient mitochondrial proteins. Mutations in both HSPA9 and GLRX5 have previously been associated with CSA. Therefore, we hypothesized that mutations in HSCB could also cause CSA. We screened patients with genetically undefined CSA and identified a frameshift mutation and a rare promoter variant in HSCB in a female patient with non-syndromic CSA. We found that HSCB expression was decreased in patient-derived fibroblasts and K562 erythroleukemia cells engineered to have the patient-specific promoter variant. Furthermore, gene knockdown and deletion experiments performed in K562 cells, zebrafish, and mice demonstrate that loss of HSCB results in impaired Fe-S cluster biogenesis, a defect in RBC hemoglobinization, and the development of siderocytes and more broadly perturbs hematopoiesis in vivo. These results further affirm the involvement of Fe-S cluster biogenesis in erythropoiesis and hematopoiesis and define HSCB as a CSA gene.


Assuntos
Anemia Sideroblástica/genética , Chaperonas Moleculares/genética , Mutação , Adolescente , Anemia Sideroblástica/congênito , Anemia Sideroblástica/metabolismo , Animais , Criança , Análise Mutacional de DNA , Feminino , Mutação da Fase de Leitura , Técnicas de Silenciamento de Genes , Humanos , Proteínas Ferro-Enxofre/deficiência , Proteínas Ferro-Enxofre/genética , Células K562 , Masculino , Camundongos , Camundongos Knockout , Chaperonas Moleculares/metabolismo , Linhagem , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas , Adulto Jovem , Peixe-Zebra
13.
Elife ; 72018 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-29745900

RESUMO

Forebrain precursor cells are dynamic during early brain development, yet the underlying molecular changes remain elusive. We observed major differences in transcriptional signatures of precursor cells from mouse forebrain at embryonic days E8.5 vs. E10.5 (before vs. after neural tube closure). Genes encoding protein biosynthetic machinery were strongly downregulated at E10.5. This was matched by decreases in ribosome biogenesis and protein synthesis, together with age-related changes in proteomic content of the adjacent fluids. Notably, c-MYC expression and mTOR pathway signaling were also decreased at E10.5, providing potential drivers for the effects on ribosome biogenesis and protein synthesis. Interference with c-MYC at E8.5 prematurely decreased ribosome biogenesis, while persistent c-MYC expression in cortical progenitors increased transcription of protein biosynthetic machinery and enhanced ribosome biogenesis, as well as enhanced progenitor proliferation leading to subsequent macrocephaly. These findings indicate large, coordinated changes in molecular machinery of forebrain precursors during early brain development.


Assuntos
Regulação para Baixo , Regulação da Expressão Gênica no Desenvolvimento , Biogênese de Organelas , Prosencéfalo/embriologia , Ribossomos/metabolismo , Animais , Camundongos , Fatores de Tempo
14.
Science ; 357(6350)2017 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-28774900

RESUMO

During terminal differentiation, the global protein complement is remodeled, as epitomized by erythrocytes, whose cytosol is ~98% globin. The erythroid proteome undergoes a rapid transition at the reticulocyte stage; however, the mechanisms driving programmed elimination of preexisting cytosolic proteins are unclear. We found that a mutation in the murine Ube2o gene, which encodes a ubiquitin-conjugating enzyme induced during erythropoiesis, results in anemia. Proteomic analysis suggested that UBE2O is a broad-spectrum ubiquitinating enzyme that remodels the erythroid proteome. In particular, ribosome elimination, a hallmark of reticulocyte differentiation, was defective in Ube2o-/- mutants. UBE2O recognized ribosomal proteins and other substrates directly, targeting them to proteasomes for degradation. Thus, in reticulocytes, the induction of ubiquitinating factors may drive the transition from a complex to a simple proteome.


Assuntos
Células Eritroides/citologia , Eritropoese/fisiologia , Proteínas Ribossômicas/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo , Ubiquitinação , Anemia/genética , Anemia Hipocrômica/genética , Animais , Eritrócitos/citologia , Eritrócitos/enzimologia , Células Eritroides/enzimologia , Eritropoese/genética , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Mutação , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteoma/metabolismo , Proteômica , Reticulócitos/citologia , Reticulócitos/enzimologia , Ribossomos/metabolismo , Enzimas de Conjugação de Ubiquitina/genética , Globinas beta/genética , Globinas beta/metabolismo
15.
Sci Rep ; 6: 26202, 2016 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-27197761

RESUMO

Mitochondrial myopathy with lactic acidosis and sideroblastic anemia (MLASA) is an oxidative phosphorylation disorder, with primary clinical manifestations of myopathic exercise intolerance and a macrocytic sideroblastic anemia. One cause of MLASA is recessive mutations in PUS1, which encodes pseudouridine (Ψ) synthase 1 (Pus1p). Here we describe a mouse model of MLASA due to mutations in PUS1. As expected, certain Ψ modifications were missing in cytoplasmic and mitochondrial tRNAs from Pus1(-/-) animals. Pus1(-/-) mice were born at the expected Mendelian frequency and were non-dysmorphic. At 14 weeks the mutants displayed reduced exercise capacity. Examination of tibialis anterior (TA) muscle morphology and histochemistry demonstrated an increase in the cross sectional area and proportion of myosin heavy chain (MHC) IIB and low succinate dehydrogenase (SDH) expressing myofibers, without a change in the size of MHC IIA positive or high SDH myofibers. Cytochrome c oxidase activity was significantly reduced in extracts from red gastrocnemius muscle from Pus1(-/-) mice. Transmission electron microscopy on red gastrocnemius muscle demonstrated that Pus1(-/-) mice also had lower intermyofibrillar mitochondrial density and smaller mitochondria. Collectively, these results suggest that alterations in muscle metabolism related to mitochondrial content and oxidative capacity may account for the reduced exercise capacity in Pus1(-/-) mice.


Assuntos
Hidroliases/deficiência , Síndrome MELAS/patologia , Músculos/patologia , Músculos/fisiologia , Animais , Modelos Animais de Doenças , Histocitoquímica , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão
16.
Am J Surg Pathol ; 38(9): 1298-304, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24618611

RESUMO

Although indolent T-lymphoblastic proliferations (iT-LBP) are rare, this diagnosis should be excluded in any patient with an extrathymic proliferation of immature TdT+T cells. Unlike T-lymphoblastic leukemia/lymphoma, patients with iT-LBP do not require chemotherapy. We report a case of iT-LBP with disseminated multinodal involvement in an otherwise healthy 49-year-old woman. Multiple lymph node biopsies were performed over the course of several months demonstrating persistent and anatomically diffuse involvement. Over 18 months, and without therapy, she has remained healthy, and her lymphadenopathy significantly improved. No bone marrow or peripheral blood involvement was ever identified. Atypical T cells showed an immunophenotypic spectrum of T-cell antigen expression with partial CD33 on a subset of T cells detected by both flow cytometry and immunohistochemistry. Both T-cell clonality and Human Androgen Receptor Assay (HUMARA) studies, performed on lymph node biopsy specimens, were negative. This case represents the first detailed clinical, morphologic, molecular, and immunophenotypic description of disseminated multinodal involvement by nonclonal iT-LBP with partial CD33 expression on T cells.


Assuntos
Biomarcadores Tumorais/análise , Proliferação de Células , Linfonodos/imunologia , Ativação Linfocitária , Transtornos Linfoproliferativos/imunologia , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico/análise , Linfócitos T/imunologia , Adulto , Biópsia , Diagnóstico Diferencial , Feminino , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Imunofenotipagem , Linfonodos/patologia , Transtornos Linfoproliferativos/patologia , Valor Preditivo dos Testes , Prognóstico , Linfócitos T/patologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA