Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Genes Dev ; 31(10): 1007-1023, 2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28611190

RESUMO

Janus kinase 2 (JAK2) is a central kinase in hematopoietic stem/progenitor cells (HSPCs), and its uncontrolled activation is a prominent oncogenic driver of hematopoietic neoplasms. However, molecular mechanisms underlying the regulation of JAK2 have remained elusive. Here we report that the Casitas B-cell lymphoma (CBL) family E3 ubiquitin ligases down-regulate JAK2 stability and signaling via the adaptor protein LNK/SH2B3. We demonstrated that depletion of CBL/CBL-B or LNK abrogated JAK2 ubiquitination, extended JAK2 half-life, and enhanced JAK2 signaling and cell growth in human cell lines as well as primary murine HSPCs. Built on these findings, we showed that JAK inhibitor (JAKi) significantly reduced aberrant HSPCs and mitigated leukemia development in a mouse model of aggressive myeloid leukemia driven by loss of Cbl and Cbl-b Importantly, primary human CBL mutated (CBLmut ) leukemias exhibited increased JAK2 protein levels and signaling and were hypersensitive to JAKi. Loss-of-function mutations in CBL E3 ubiquitin ligases are found in a wide range of myeloid malignancies, which are diseases without effective treatment options. Hence, our studies reveal a novel signaling axis that regulates JAK2 in normal and malignant HSPCs and suggest new therapeutic strategies for treating CBLmut myeloid malignancies.


Assuntos
Janus Quinase 2/metabolismo , Leucemia Mieloide Aguda/enzimologia , Leucemia Mieloide Aguda/fisiopatologia , Proteínas Proto-Oncogênicas c-cbl/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Linhagem Celular Tumoral , Proliferação de Células/genética , Citocinas/metabolismo , Estabilidade Enzimática , Células-Tronco Hematopoéticas/enzimologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Janus Quinase 2/genética , Leucemia Mieloide Aguda/genética , Proteínas de Membrana , Camundongos , Mutação , Proteólise , Proteínas Proto-Oncogênicas c-cbl/genética , Transdução de Sinais/genética , Ubiquitinação
2.
J Cutan Pathol ; 49(10): 885-888, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35708461

RESUMO

Lafora disease is a rare inherited neurodegenerative disease with onset in adolescence. Patients present with progressive myoclonic seizures and cognitive decline. The disease is linked to mutations in either of the two genes encoding malin and laforin, and it is associated with the accumulation of polyglucosan inclusions (Lafora bodies [LBs]) in various tissues, such as brain, liver, muscle, and skin, with the skin being particularly accessible for biopsy. Histopathologic examination of affected tissue with demonstration of LBs, together with the presence of pathologic mutation in EPM2A or NHLRC1 genes, is sufficient for diagnosis of this neurologic disorder when clinically suspected. Here, we report the case of a 16-year-old female with progressive neurologic symptoms and homozygous mutation in the NHLRC1 gene encoding malin. The skin biopsy was instrumental in reaching the final diagnosis by showing LBs in sweat glands by histopathologic and electron microscopic examination.


Assuntos
Doença de Lafora , Doenças Neurodegenerativas , Adolescente , Biópsia , Proteínas de Transporte/genética , Feminino , Humanos , Doença de Lafora/diagnóstico , Doença de Lafora/genética , Doença de Lafora/patologia , Proteínas Tirosina Fosfatases não Receptoras/genética , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
3.
Blood ; 133(14): 1560-1571, 2019 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-30755420

RESUMO

Hematopoietic stem cell (HSC) homeostasis is controlled by cytokine receptor-mediated Janus kinase 2 (JAK2) signaling. We previously found that JAK2 is promptly ubiquitinated upon cytokine stimulation. Whether a competing JAK2 deubiquitination activity exists is unknown. LNK is an essential adaptor protein that constrains HSC expansion through dampening thrombopoietin (TPO)-induced JAK2 signaling. We show here that a LNK-associated lysine-63 (K63)-deubiquitinating enzyme complex, Brcc36 isopeptidase complex (BRISC), attenuates HSC expansion through control of JAK2 signaling. We pinpoint a direct interaction between the LNK SH2 domain and a phosphorylated tyrosine residue in KIAA0157 (Abraxas2), a unique and defining BRISC component. Kiaa0157 deficiency in mice led to an expansion of phenotypic and functional HSCs. Endogenous JAK2 and phospho-JAK2 were rapidly K63-ubiquitinated upon TPO stimulation, and this action was augmented in cells depleted of the BRISC core components KIAA0157, MERIT40, or BRCC36. This increase in JAK2 ubiquitination after BRISC knockdown was associated with increased TPO-mediated JAK2 activation and protein levels, and increased MPL receptor presence at the cell surface. In addition, BRISC depletion promoted membrane proximal association between the MPL receptor and pJAK2/JAK2, thus enhancing activated JAK2/MPL at the cell membrane. These findings define a novel pathway by which K63-ubiquitination promotes JAK2 stability and activation in a proteasome-independent manner. Moreover, mutations in BRCC36 are found in clonal hematopoiesis in humans. This research may shed light on the mechanistic understanding of a potential role of BRCC36 in human HSCs.


Assuntos
Proliferação de Células , Enzimas Desubiquitinantes/fisiologia , Células-Tronco Hematopoéticas/citologia , Janus Quinase 2/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Enzimas Desubiquitinantes/genética , Humanos , Camundongos , Proteínas Associadas à Matriz Nuclear/metabolismo , Receptores de Trombopoetina/metabolismo , Transdução de Sinais , Trombopoetina/farmacologia , Proteases Específicas de Ubiquitina/metabolismo , Ubiquitinação , Domínios de Homologia de src
6.
Blood ; 125(11): 1730-8, 2015 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-25636339

RESUMO

Hematopoietic stem cell (HSC) self-renewal and multilineage reconstitution are controlled by positive and negative signaling cues with perturbations leading to disease. Lnk is an essential signaling adaptor protein that dampens signaling by the cytokine thrombopoietin (Tpo) to limit HSC expansion. Here, we show that MERIT40 (Mediator of RAP80 Interactions and Targeting 40 kDa [M40]), a core subunit of an Lnk-associated Lys63 deubiquitinating (DUB) complex, attenuates HSC expansion. M40 deficiency increases the size of phenotypic and functional HSC pools. M40(-/-) HSCs are more resistant to cytoablative stress, and exhibit superior repopulating ability and self-renewal upon serial transplantation. M40(-/-) HSCs display increased quiescence and decelerated cell cycle kinetics accompanied by downregulation of gene sets associated with cell division. Mechanistically, M40 deficiency triggers hypersensitivity to Tpo stimulation and the stem cell phenotypes are abrogated on a background null for the Tpo receptor Mpl. These results establish M40-containing DUB complexes as novel HSC regulators of HSC expansion, implicate Lys63 ubiquitination in HSC signaling, and point to DUB-specific inhibitors as reagents to expand stem cell populations.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/deficiência , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Receptores de Trombopoetina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Ciclo Celular , Proliferação de Células , Ensaio de Unidades Formadoras de Colônias , Feminino , Fluoruracila/toxicidade , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Trombopoetina/deficiência , Receptores de Trombopoetina/genética , Transdução de Sinais , Estresse Fisiológico , Trombopoetina/metabolismo , Proteases Específicas de Ubiquitina/metabolismo
7.
J Appl Lab Med ; 8(4): 742-750, 2023 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-37232112

RESUMO

BACKGROUND: Multiple laboratory tests are employed for detection of monoclonal proteins in patients and include serum protein electrophoresis (SPEP), immunofixation electrophoresis, free light chain (FLC) immunoassay, and mass spectrometry (Mass-Fix). Recently, reports on a drift in FLC quantitation results have been brought to light. METHODS: We studied a cohort of 16 887 patients whose sera were tested for a monoclonal protein by a FLC assay, serum protein electrophoresis, and Mass-Fix. This is a retrospective study designed to assess the impact of a drift on the performance of FLC ratio (rFLC) in groups of patients with and without detectable plasma cell disorders (PCDs). RESULTS: The results demonstrated that 63% of patients with monoclonal protein equal or higher than 2 g/L (by SPEP) had an abnormal rFLC (reference range 0.26-1.65). Conversely, 16% of patients with undetectable monoclonal protein by other methods (i.e., SPEP and Mass-Fix) who also had no record of treated PCD had an abnormal rFLC. In these cases, there was an imbalance in the number of kappa high rFLCs to lambda low rFLCs of 201 to 1. CONCLUSIONS: The results of this study suggest decreased specificity of rFLC for a monoclonal kappa FLC in the 1.65 to 3.0 range.


Assuntos
Gamopatia Monoclonal de Significância Indeterminada , Paraproteinemias , Humanos , Estudos Retrospectivos , Cadeias lambda de Imunoglobulina , Sensibilidade e Especificidade , Paraproteinemias/diagnóstico , Cadeias Leves de Imunoglobulina , Cadeias kappa de Imunoglobulina , Gamopatia Monoclonal de Significância Indeterminada/diagnóstico , Anticorpos Monoclonais
8.
J Biol Chem ; 285(27): 21103-13, 2010 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-20236926

RESUMO

Acid sphingomyelinase (ASMase) has been proposed to mediate lipopolysaccharide (LPS) signaling in various cell types. This study shows that ASMase is a negative regulator of LPS-induced tumor necrosis factor alpha (TNFalpha) secretion in macrophages. ASMase-deficient (asm(-/-)) mice and isolated peritoneal macrophages produce severalfold more TNFalpha than their wild-type (asm(+/+)) counterparts when stimulated with LPS, whereas the addition of exogenous ceramides or sphingomyelinase reduces the differences. The underlying mechanism for these effects is not transcriptional but post-translational. The TNFalpha-converting enzyme (TACE) catalyzes the maturation of the 26-kDa precursor (pro-TNFalpha) to an active 17-kDa form (soluble (s)TNFalpha). In mouse peritoneal macrophages, the activity of TACE was the rate-limiting factor regulating TNFalpha production. A substantial portion of the translated pro-TNFalpha was not processed to sTNFalpha; instead, it was rapidly internalized and degraded in the lysosomes. TACE activity was 2-3-fold higher in asm(-/-) macrophages as compared with asm(+/+) macrophages and was suppressed when cells were treated with exogenous ceramide and sphingomyelinase. Indirect immunofluorescence analyses revealed distinct TNFalpha-positive structures in the close vicinity of the plasma membrane in asm(-/-) but not in asm(+/+) macrophages. asm(-/-) cells also had a higher number of early endosomal antigen 1-positive early endosomes. Experiments that involved inhibitors of TACE, endocytosis, and lysosomal proteolysis suggest that in the asm(-/-) cells a significant portion of pro-TNFalpha was sequestered within the early endosomes, and instead of undergoing lysosomal proteolysis, it was recycled to the plasma membrane and processed to sTNFalpha.


Assuntos
Proteínas ADAM/metabolismo , Ceramidas/metabolismo , Macrófagos Peritoneais/fisiologia , Esfingomielina Fosfodiesterase/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteína ADAM17 , Animais , Ensaio de Imunoadsorção Enzimática , Técnica Indireta de Fluorescência para Anticorpo , Genótipo , Heterozigoto , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Cinética , Lipopolissacarídeos/farmacologia , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/enzimologia , Macrófagos Peritoneais/metabolismo , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase/métodos , RNA/genética , RNA/isolamento & purificação , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Esfingomielina Fosfodiesterase/deficiência , Esfingomielina Fosfodiesterase/genética , Fator de Necrose Tumoral alfa/genética
9.
Hum Pathol ; 114: 66-73, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34019867

RESUMO

T-lymphoblastic leukemia/lymphoma (T-ALL/LBL) is a rare acute leukemia that expresses cytoplasmic CD3 (cCD3) and frequently lacks surface CD3. Given that routine flow cytometric testing for cCD3 may not be feasible and cCD3 interpretation may be difficult, we investigate if surface CD2 and/or CD7 expression on blasts can be used by flow cytometry to screen for T-lineage acute leukemia. We retrospectively reviewed flow cytometric data from 233 acute leukemias (36 T-ALL/LBL, 8 mixed-phenotype acute leukemia T/myeloid, 80 acute myeloid leukemia, 97 B-ALL/LBL, 8 mixed-phenotype acute leukemia B/myeloid, and 4 acute undifferentiated leukemia cases). Uniform expression (≥75% of blasts) of CD2 and/or CD7 was seen in all 44 cCD3-positive cases but in only 11% (20/189) of cCD3-negative acute leukemias, thus demonstrating 100% sensitivity and 89% specificity in the identification of cCD3-positive (T-lineage) acute leukemia. To avoid selection bias, we prospectively studied 232 consecutive acute leukemias for which cCD3, CD2, and CD7 were automatically performed in all cases. Similar to the retrospective study, uniform expression of CD2 and/or CD7 on blasts showed 100% sensitivity and 88% specificity in the screening for cCD3-positive (T-lineage) acute leukemia. Therefore, acute leukemias with uniform expression of CD2 and/or CD7 warrant further testing for cCD3 to evaluate for T-lineage acute leukemia. Blasts that lack both uniform CD2 and CD7 expression do not require additional cCD3 testing. We propose that CD2 and CD7 could be utilized in a limited antibody flow cytometry panel as a sensitive, robust, and cost-effective way to screen for T-lineage acute leukemia.


Assuntos
Antígenos CD7/análise , Biomarcadores Tumorais/análise , Antígenos CD2/análise , Linhagem da Célula , Citometria de Fluxo , Imunofenotipagem , Leucemia-Linfoma Linfoblástico de Células T Precursoras/imunologia , Linfócitos T/imunologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Complexo CD3/análise , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Fenótipo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patologia , Valor Preditivo dos Testes , Estudos Prospectivos , Reprodutibilidade dos Testes , Estudos Retrospectivos , Adulto Jovem
10.
Nat Commun ; 9(1): 3915, 2018 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-30254368

RESUMO

Fanconi anemia (FA) is a bone marrow failure (BMF) syndrome that arises from mutations in a network of FA genes essential for DNA interstrand crosslink (ICL) repair and replication stress tolerance. While allogeneic stem cell transplantation can replace defective HSCs, interventions to mitigate HSC defects in FA do not exist. Remarkably, we reveal here that Lnk (Sh2b3) deficiency restores HSC function in Fancd2-/- mice. Lnk deficiency does not impact ICL repair, but instead stabilizes stalled replication forks in a manner, in part, dependent upon alleviating blocks to cytokine-mediated JAK2 signaling. Lnk deficiency restores proliferation and survival of Fancd2-/- HSCs, while reducing replication stress and genomic instability. Furthermore, deletion of LNK in human FA-like HSCs promotes clonogenic growth. These findings highlight a new role for cytokine/JAK signaling in promoting replication fork stability, illuminate replication stress as a major underlying origin of BMF in FA, and have strong therapeutic implications.


Assuntos
Proteína do Grupo de Complementação D2 da Anemia de Fanconi/genética , Anemia de Fanconi/genética , Instabilidade Genômica/genética , Células-Tronco Hematopoéticas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas Adaptadoras de Transdução de Sinal , Animais , Transplante de Medula Óssea , Proliferação de Células/genética , Células Cultivadas , Reparo do DNA/genética , Replicação do DNA/genética , Anemia de Fanconi/metabolismo , Anemia de Fanconi/terapia , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/deficiência , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Masculino , Proteínas de Membrana , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout
13.
Aging Cell ; 11(6): 949-59, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22812478

RESUMO

Upon aging, the number of hematopoietic stem cells (HSCs) in the bone marrow increases while their repopulation potential declines. Moreover, aged HSCs exhibit lineage bias in reconstitution experiments with an inclination toward myeloid at the expense of lymphoid potential. The adaptor protein Lnk is an important negative regulator of HSC homeostasis, as Lnk deficiency is associated with a 10-fold increase in HSC numbers in young mice. However, the age-related increase in functional HSC numbers found in wild-type HSCs was not observed in Lnk-deficient animals. Importantly, HSCs from aged Lnk null mice possess greatly enhanced self-renewal capacity and diminished exhaustion, as evidenced by serial transplant experiments. In addition, Lnk deficiency ameliorates the aging-associated lineage bias. Transcriptome analysis revealed that WT and Lnk-deficient HSCs share many aging-related changes in gene expression patterns. Nonetheless, Lnk null HSCs displayed altered expression of components in select signaling pathways with potential involvement in HSC self-renewal and aging. Taken together, these results suggest that loss of Lnk partially mitigates age-related HSC alterations.


Assuntos
Envelhecimento/genética , Linhagem da Célula/genética , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Hematopoéticas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas Adaptadoras de Transdução de Sinal , Adulto , Animais , Contagem de Células , Proliferação de Células , Citometria de Fluxo , Perfilação da Expressão Gênica , Genótipo , Células-Tronco Hematopoéticas/citologia , Humanos , Proteínas de Membrana , Camundongos , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Transdução de Sinais , Transcriptoma
14.
J Clin Invest ; 122(6): 2079-91, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22546852

RESUMO

Hematopoietic stem and progenitor cell (HSPC) functions are governed by intricate signaling networks. The tyrosine kinase JAK2 plays an essential role in cytokine signaling during hematopoiesis. The adaptor protein LNK is a critical determinant of this process through its inhibitory interaction with JAK2, thereby limiting HSPC self-renewal. LNK deficiency promotes myeloproliferative neoplasm (MPN) development in mice, and LNK loss-of-function mutations are found in human MPNs, emphasizing its pivotal role in normal and malignant HSPCs. Here, we report the identification of 14-3-3 proteins as LNK binding partners. 14-3-3 interfered with the LNK-JAK2 interaction, thereby alleviating LNK inhibition of JAK2 signaling and cell proliferation. Binding of 14-3-3 required 2 previously unappreciated serine phosphorylation sites in LNK, and we found that their phosphorylation is mediated by glycogen synthase kinase 3 and PKA kinases. Mutations of these residues abrogated the interaction and augmented the growth inhibitory function of LNK. Conversely, forced 14-3-3 binding constrained LNK function. Furthermore, interaction with 14-3-3 sequestered LNK in the cytoplasm away from the plasma membrane-proximal JAK2. Importantly, bone marrow transplantation studies revealed an essential role for 14-3-3 in HSPC reconstitution that can be partially mitigated by LNK deficiency. We believe that, together, this work implicates 14-3-3 proteins as novel and positive HSPC regulators by impinging on the LNK/JAK2 pathway.


Assuntos
Proteínas 14-3-3/metabolismo , Proliferação de Células , Células-Tronco Hematopoéticas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Janus Quinase 2/metabolismo , Transdução de Sinais , Proteínas 14-3-3/genética , Proteínas Adaptadoras de Transdução de Sinal , Animais , Transplante de Medula Óssea , Proteínas Quinases Dependentes de AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Janus Quinase 2/genética , Proteínas de Membrana , Camundongos , Camundongos Knockout , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/metabolismo , Fosforilação/genética , Transplante Homólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA