Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Blood ; 142(3): 244-259, 2023 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-37084382

RESUMO

Vacuoles, E1 enzyme, X-linked, autoinflammatory, somatic (VEXAS) syndrome is caused by somatic mutations in UBA1 (UBA1mut) and characterized by heterogenous systemic autoinflammation and progressive hematologic manifestations, meeting criteria for myelodysplastic syndrome (MDS) and plasma cell dyscrasias. The landscape of myeloid-related gene mutations leading to typical clonal hematopoiesis (CH) in these patients is unknown. Retrospectively, we screened 80 patients with VEXAS for CH in their peripheral blood (PB) and correlated the findings with clinical outcomes in 77 of them. UBA1mut were most common at hot spot p.M41 (median variant allele frequency [VAF] = 75%). Typical CH mutations cooccurred with UBA1mut in 60% of patients, mostly in DNMT3A and TET2, and were not associated with inflammatory or hematologic manifestations. In prospective single-cell proteogenomic sequencing (scDNA), UBA1mut was the dominant clone, present mostly in branched clonal trajectories. Based on integrated bulk and scDNA analyses, clonality in VEXAS followed 2 major patterns: with either typical CH preceding UBA1mut selection in a clone (pattern 1) or occurring as an UBA1mut subclone or in independent clones (pattern 2). VAF in the PB differed markedly between DNMT3A and TET2 clones (median VAF of 25% vs 1%). DNMT3A and TET2 clones associated with hierarchies representing patterns 1 and 2, respectively. Overall survival for all patients was 60% at 10 years. Transfusion-dependent anemia, moderate thrombocytopenia, and typical CH mutations, each correlated with poor outcome. In VEXAS, UBA1mut cells are the primary cause of systemic inflammation and marrow failure, being a new molecularly defined somatic entity associated with MDS. VEXAS-associated MDS is distinct from classical MDS in its presentation and clinical course.


Assuntos
Hematopoiese Clonal , Dermatite , Humanos , Hematopoiese Clonal/genética , Estudos Prospectivos , Estudos Retrospectivos , Mutação
2.
Ann Rheum Dis ; 83(4): 508-517, 2024 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-38049983

RESUMO

OBJECTIVES: Ageing and inflammation are associated with clonal haematopoiesis (CH), the emergence of somatic mutations in haematopoietic cells. This study details CH in patients with systemic vasculitis in association with clinical, haematological and immunological parameters. METHODS: Patients with three forms of vasculitis were screened for CH in peripheral blood by error-corrected sequencing. Relative contributions of age and vasculitis on CH prevalence were calculated using multivariable logistic regression. Clonal hierarchies were assessed by proteogenomic single-cell DNA sequencing, and functional experiments were performed in association with CH status. RESULTS: Patients with Takayasu's arteritis (TAK; n=70; mean age=33.2 years), antineutrophil cytoplasmic antibody-associated vasculitis (AAV; n=47; mean age=55.3 years) and giant cell arteritis (GCA; n=59; mean age=71.2 years) were studied. CH, most commonly in DNMT3A and TET2, was detected in 34% (60/176) of patients versus 18% (28/151) of age-matched controls (p<0.01). Prevalence of CH was independently associated with age (standardised B=0.96, p<0.01) and vasculitis (standardised B=0.46, p<0.01), occurring in 61%, 32% and 13% of patients with GCA, AAV and TAK, respectively. Both branched and linear clonal trajectories showed myeloid-lineage bias, and CH was associated with markers of cellular activation. In GCA, mutations were detected in temporal artery biopsies, and clinical relapse correlated with CH in a dose-dependent relationship with clone size. CONCLUSIONS: Age was more strongly associated with CH prevalence than inflammation in systemic vasculitis. Clonal profile was dominated by DNMT3A mutations which were associated with relapse in GCA. CH is not likely a primary causal factor in systemic vasculitis but may contribute to inflammation.


Assuntos
Vasculite Associada a Anticorpo Anticitoplasma de Neutrófilos , Arterite de Células Gigantes , Arterite de Takayasu , Humanos , Adulto , Pessoa de Meia-Idade , Idoso , Arterite de Células Gigantes/epidemiologia , Arterite de Takayasu/epidemiologia , Hematopoiese Clonal , Inflamação , Recidiva
3.
Blood ; 138(26): 2799-2809, 2021 12 30.
Artigo em Inglês | MEDLINE | ID: mdl-34724566

RESUMO

Immune aplastic anemia (AA) features somatic loss of HLA class I allele expression on bone marrow cells, consistent with a mechanism of escape from T-cell-mediated destruction of hematopoietic stem and progenitor cells. The clinical significance of HLA abnormalities has not been well characterized. We examined the somatic loss of HLA class I alleles and correlated HLA loss and mutation-associated HLA genotypes with clinical presentation and outcomes after immunosuppressive therapy in 544 AA patients. HLA class I allele loss was detected in 92 (22%) of the 412 patients tested, in whom there were 393 somatic HLA gene mutations and 40 instances of loss of heterozygosity. Most frequently affected was HLA-B*14:02, followed by HLA-A*02:01, HLA-B*40:02, HLA-B*08:01, and HLA-B*07:02. HLA-B*14:02, HLA-B*40:02, and HLA-B*07:02 were also overrepresented in AA. High-risk clonal evolution was correlated with HLA loss, HLA-B*14:02 genotype, and older age, which yielded a valid prediction model. In 2 patients, we traced monosomy 7 clonal evolution from preexisting clones harboring somatic mutations in HLA-A*02:01 and HLA-B*40:02. Loss of HLA-B*40:02 correlated with higher blood counts. HLA-B*07:02 and HLA-B*40:01 genotypes and their loss correlated with late-onset of AA. Our results suggest the presence of specific immune mechanisms of molecular pathogenesis with clinical implications. HLA genotyping and screening for HLA loss may be of value in the management of immune AA. This study was registered at clinicaltrials.gov as NCT00001964, NCT00061360, NCT00195624, NCT00260689, NCT00944749, NCT01193283, and NCT01623167.


Assuntos
Anemia Aplástica/genética , Genes MHC Classe I , Antígenos HLA-A/genética , Antígenos HLA-B/genética , Mutação , Adolescente , Adulto , Alelos , Anemia Aplástica/imunologia , Evolução Clonal , Feminino , Deleção de Genes , Expressão Gênica , Antígenos HLA-A/imunologia , Antígenos HLA-B/imunologia , Humanos , Imunidade , Perda de Heterozigosidade , Masculino , Pessoa de Meia-Idade , Adulto Jovem
4.
Blood ; 136(21): 2373-2385, 2020 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-32929473

RESUMO

Inversion of chromosome 16 is a consistent finding in patients with acute myeloid leukemia subtype M4 with eosinophilia, which generates a CBFB-MYH11 fusion gene. It is generally considered that CBFß-SMMHC, the fusion protein encoded by CBFB-MYH11, is a dominant negative repressor of RUNX1. However, recent findings challenge the RUNX1-repression model for CBFß-SMMHC-mediated leukemogenesis. To definitively address the role of Runx1 in CBFB-MYH11-induced leukemia, we crossed conditional Runx1 knockout mice (Runx1f/f) with conditional Cbfb-MYH11 knockin mice (Cbfb+/56M). On Mx1-Cre activation in hematopoietic cells induced by poly (I:C) injection, all Mx1-CreCbfb+/56M mice developed leukemia in 5 months, whereas no leukemia developed in Runx1f/fMx1-CreCbfb+/56M mice, and this effect was cell autonomous. Importantly, the abnormal myeloid progenitors (AMPs), a leukemia-initiating cell population induced by Cbfb-MYH11 in the bone marrow, decreased and disappeared in Runx1f/fMx1-CreCbfb+/56M mice. RNA-seq analysis of AMP cells showed that genes associated with proliferation, differentiation blockage, and leukemia initiation were differentially expressed between Mx1-CreCbfb+/56M and Runx1f/fMx1-CreCbfb+/56M mice. In addition, with the chromatin immunocleavage sequencing assay, we observed a significant enrichment of RUNX1/CBFß-SMMHC target genes in Runx1f/fMx1-CreCbfb+/56M cells, especially among downregulated genes, suggesting that RUNX1 and CBFß-SMMHC mainly function together as activators of gene expression through direct target gene binding. These data indicate that Runx1 is indispensable for Cbfb-MYH11-induced leukemogenesis by working together with CBFß-SMMHC to regulate critical genes associated with the generation of a functional AMP population.


Assuntos
Transformação Celular Neoplásica/genética , Subunidade alfa 2 de Fator de Ligação ao Core/fisiologia , Regulação Leucêmica da Expressão Gênica , Leucemia Experimental/genética , Células Mieloides/metabolismo , Proteínas de Neoplasias/fisiologia , Células-Tronco Neoplásicas/metabolismo , Proteínas de Fusão Oncogênica/fisiologia , Ativação Transcricional , Animais , Subunidade alfa 2 de Fator de Ligação ao Core/deficiência , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Técnicas de Introdução de Genes , Humanos , Leucemia Experimental/etiologia , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/citologia , Células-Tronco Neoplásicas/citologia , Proteínas de Fusão Oncogênica/genética , Poli I-C/farmacologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Neoplásico/biossíntese , RNA Neoplásico/genética , RNA-Seq , Análise de Célula Única
5.
Br J Haematol ; 190(4): 610-617, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32311088

RESUMO

Immune aplastic anaemia (AA) is caused by cytotoxic T lymphocytes (CTLs) that destroy haematopoietic stem and progenitor cells. Enhanced type 1 T helper (Th1) responses and reduced regulatory T cells (Tregs) are involved in the immune pathophysiology. CD24hi CD38hi regulatory B cells (Bregs) suppress CTLs and Th1 responses, and induce Tregs via interleukin 10 (IL-10). We investigated circulating B-cell subpopulations, including CD24hi CD38hi Bregs, as well as total B cells, CD4+ T cells, CD8+ T cells and natural killer cells in 104 untreated patients with severe and very severe AA, aged ≥18 years. All patients were treated with standard immunosuppressive therapy (IST) plus eltrombopag. CD24hi CD38hi Bregs were markedly reduced in patients with AA compared to healthy individuals, especially in very severe AA, but residual Bregs remained functional, capable of producing IL-10; total B-cell counts and the other B-cell subpopulations were similar to those of healthy individuals. CD24hi CD38hi Bregs did not correlate with responses to IST, and they recovered to levels present in healthy individuals after therapy. Mature naïve B-cell counts were unexpectedly associated with IST response. Markedly reduced CD24hi CD38hi Bregs, especially in very severe AA, with recovery after IST suggest Breg deficits may contribute to the pathophysiology of immune AA.


Assuntos
ADP-Ribosil Ciclase 1/análise , Anemia Aplástica/sangue , Antígenos CD19/análise , Subpopulações de Linfócitos B/patologia , Linfócitos B Reguladores/patologia , Antígeno CD24/análise , Linfopenia/etiologia , Glicoproteínas de Membrana/análise , Adolescente , Adulto , Idoso , Anemia Aplástica/complicações , Anemia Aplástica/tratamento farmacológico , Anemia Aplástica/patologia , Soro Antilinfocitário/uso terapêutico , Subpopulações de Linfócitos B/química , Linfócitos B Reguladores/química , Benzoatos/uso terapêutico , Medula Óssea/patologia , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD8-Positivos/patologia , Ciclosporina/uso terapêutico , Feminino , Humanos , Hidrazinas/uso terapêutico , Imunossupressores/uso terapêutico , Interleucina-10/biossíntese , Interleucina-10/genética , Células Matadoras Naturais/patologia , Contagem de Linfócitos , Linfopenia/sangue , Linfopenia/patologia , Masculino , Pessoa de Meia-Idade , Pirazóis/uso terapêutico , Receptores de Trombopoetina/agonistas , Adulto Jovem
6.
Blood ; 130(22): 2431-2442, 2017 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-29018080

RESUMO

Inversion of chromosome 16 is a consistent finding in patients with acute myeloid leukemia subtype M4 with eosinophilia, which generates a CBFB-MYH11 fusion gene. Previous studies showed that the interaction between CBFß-smooth muscle myosin heavy chain (SMMHC; encoded by CBFB-MYH11) and RUNX1 plays a critical role in the pathogenesis of this leukemia. Recently, it was shown that chromodomain helicase DNA-binding protein-7 (CHD7) interacts with RUNX1 and suppresses RUNX1-induced expansion of hematopoietic stem and progenitor cells. These results suggest that CHD7 is also critical for CBFB-MYH11-induced leukemogenesis. To test this hypothesis, we generated Chd7f/fMx1-CreCbfb+/56M mice, which expressed the Cbfb-MYH11 fusion gene and deactivated Chd7 in hematopoietic cells upon inducing Cre with polyinosinic-polycytidylic acid. The Lin-Sca1-c-Kit+ (LK) population was significantly lower in Chd7f/fMx1-CreCbfb+/56M mice than in Mx1-CreCbfb+/56M mice. In addition, there were fewer 5-bromo-2'-deoxyuridine-positive cells in the LK population in Chd7f/fMx1-CreCbfb+/56M mice, and genes associated with cell cycle, cell growth, and proliferation were differentially expressed between Chd7f/fMx1-CreCbfb+/56M and Mx1-CreCbfb+/56M leukemic cells. In vitro studies showed that CHD7 interacted with CBFß-SMMHC through RUNX1 and that CHD7 enhanced transcriptional activity of RUNX1 and CBFß-SMMHC on Csf1r, a RUNX1 target gene. Moreover, RNA sequencing of c-Kit+ cells showed that CHD7 functions mostly through altering the expression of RUNX1 target genes. Most importantly, Chd7 deficiency delayed Cbfb-MYH11-induced leukemia in both primary and transplanted mice. These data indicate that Chd7 is important for Cbfb-MYH11-induced leukemogenesis by facilitating RUNX1 regulation of transcription and cellular proliferation.


Assuntos
Proteínas de Ligação a DNA/genética , Regulação Leucêmica da Expressão Gênica , Leucemia/genética , Proteínas de Fusão Oncogênica/genética , Animais , Ciclo Celular , Proliferação de Células , Deleção de Genes , Humanos , Leucemia/patologia , Camundongos , Camundongos Knockout
7.
Blood ; 121(4): 638-42, 2013 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-23152542

RESUMO

The C-terminus of CBFß-SMMHC, the fusion protein produced by a chromosome 16 inversion in acute myeloid leukemia subtype M4Eo, contains domains for self-multimerization and transcriptional repression, both of which have been proposed to be important for leukemogenesis by CBFß-SMMHC. To test the role of the fusion protein's C-terminus in vivo, we generated knock-in mice expressing a C-terminally truncated CBFß-SMMHC (CBFß-SMMHCΔC95). Embryos with a single copy of CBFß-SMMHCΔC95 were viable and showed no defects in hematopoiesis, whereas embryos homozygous for the CBFß-SMMHCΔC95 allele had hematopoietic defects and died in mid-gestation, similar to embryos with a single-copy of the full-length CBFß-SMMHC. Importantly, unlike mice expressing full-length CBFß-SMMHC, none of the mice expressing CBFß-SMMHCΔC95 developed leukemia, even after treatment with a mutagen, although some of the older mice developed a nontransplantable myeloproliferative disease. Our data indicate that the CBFß-SMMHC's C-terminus is essential to induce embryonic hematopoietic defects and leukemogenesis.


Assuntos
Transformação Celular Neoplásica/genética , Hematopoese/genética , Leucemia/genética , Proteínas de Fusão Oncogênica/genética , Animais , Transformação Celular Neoplásica/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Regulação Leucêmica da Expressão Gênica , Técnicas de Introdução de Genes , Humanos , Camundongos , Camundongos Transgênicos , Proteínas de Fusão Oncogênica/química , Proteínas de Fusão Oncogênica/metabolismo
8.
Proc Natl Acad Sci U S A ; 109(36): 14592-7, 2012 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-22912405

RESUMO

Core binding factor (CBF) leukemias, those with translocations or inversions that affect transcription factor genes RUNX1 or CBFB, account for ~24% of adult acute myeloid leukemia (AML) and 25% of pediatric acute lymphocytic leukemia (ALL). Current treatments for CBF leukemias are associated with significant morbidity and mortality, with a 5-y survival rate of ~50%. We hypothesize that the interaction between RUNX1 and CBFß is critical for CBF leukemia and can be targeted for drug development. We developed high-throughput AlphaScreen and time-resolved fluorescence resonance energy transfer (TR-FRET) methods to quantify the RUNX1-CBFß interaction and screen a library collection of 243,398 compounds. Ro5-3335, a benzodiazepine identified from the screen, was able to interact with RUNX1 and CBFß directly, repress RUNX1/CBFB-dependent transactivation in reporter assays, and repress runx1-dependent hematopoiesis in zebrafish embryos. Ro5-3335 preferentially killed human CBF leukemia cell lines, rescued preleukemic phenotype in a RUNX1-ETO transgenic zebrafish, and reduced leukemia burden in a mouse CBFB-MYH11 leukemia model. Our data thus confirmed that RUNX1-CBFß interaction can be targeted for leukemia treatment and we have identified a promising lead compound for this purpose.


Assuntos
Benzodiazepinas/farmacologia , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Subunidade beta de Fator de Ligação ao Core/metabolismo , Ensaios de Triagem em Larga Escala/métodos , Leucemia Mieloide Aguda/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Ativação Transcricional/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Western Blotting , Subunidade beta de Fator de Ligação ao Core/genética , Ensaio de Desvio de Mobilidade Eletroforética , Citometria de Fluxo , Transferência Ressonante de Energia de Fluorescência/métodos , Vetores Genéticos/genética , Hematopoese/efeitos dos fármacos , Técnicas Histológicas , Humanos , Imunoprecipitação , Células Jurkat , Camundongos , Dados de Sequência Molecular , Mapeamento de Interação de Proteínas/métodos , Ressonância de Plasmônio de Superfície , Peixe-Zebra
9.
Blood ; 119(6): 1511-21, 2012 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-22160378

RESUMO

KIT mutations are the most common secondary mutations in inv(16) acute myeloid leukemia (AML) patients and are associated with poor prognosis. It is therefore important to verify that KIT mutations cooperate with CBFB-MYH11, the fusion gene generated by inv(16), for leukemogenesis. Here, we transduced wild-type and conditional Cbfb-MYH11 knockin (KI) mouse bone marrow (BM) cells with KIT D816V/Y mutations. KIT transduction caused massive BM Lin(-) cell death and fewer colonies in culture that were less severe in the KI cells. D816Y KIT but not wild-type KIT enhanced proliferation in Lin(-) cells and led to more mixed lineage colonies from transduced KI BM cells. Importantly, 60% and 80% of mice transplanted with KI BM cells expressing D816V or D816Y KIT, respectively, died from leukemia within 9 months, whereas no control mice died. Results from limiting dilution transplantations indicate higher frequencies of leukemia-initiating cells in the leukemia expressing mutated KIT. Signaling pathway analysis revealed that p44/42 MAPK and Stat3, but not AKT and Stat5, were strongly phosphorylated in the leukemia cells. Finally, leukemia cells carrying KIT D816 mutations were sensitive to the kinase inhibitor PKC412. Our data provide clear evidence for cooperation between mutated KIT and CBFB-MYH11 during leukemogenesis.


Assuntos
Leucemia/genética , Mutação , Proteínas de Fusão Oncogênica/genética , Proteínas Proto-Oncogênicas c-kit/genética , Animais , Western Blotting , Transplante de Medula Óssea , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Progressão da Doença , Feminino , Citometria de Fluxo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Leucemia/metabolismo , Leucemia/patologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais , Estaurosporina/análogos & derivados , Estaurosporina/farmacologia
10.
Cell Rep Med ; 4(8): 101160, 2023 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-37586319

RESUMO

VEXAS (vacuoles, E1 enzyme, X-linked, autoinflammatory, somatic) syndrome is a pleiotropic, severe autoinflammatory disease caused by somatic mutations in the ubiquitin-like modifier activating enzyme 1 (UBA1) gene. To elucidate VEXAS pathophysiology, we performed transcriptome sequencing of single bone marrow mononuclear cells and hematopoietic stem and progenitor cells (HSPCs) from VEXAS patients. HSPCs are biased toward myeloid (granulocytic) differentiation, and against lymphoid differentiation in VEXAS. Activation of multiple inflammatory pathways (interferons and tumor necrosis factor alpha) occurs ontogenically early in primitive hematopoietic cells and particularly in the myeloid lineage in VEXAS, and inflammation is prominent in UBA1-mutated cells. Dysregulation in protein degradation likely leads to higher stress response in VEXAS HSPCs, which positively correlates with inflammation. TCR usage is restricted and there are increased cytotoxicity and IFN-γ signaling in T cells. In VEXAS syndrome, both aberrant inflammation and myeloid predominance appear intrinsic to hematopoietic stem cells mutated in UBA1.


Assuntos
Células-Tronco Hematopoéticas , Inflamação , Humanos , Proteólise , Diferenciação Celular , Inflamação/genética
11.
Blood ; 115(7): 1433-43, 2010 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-20007544

RESUMO

It is known that CBFB-MYH11, the fusion gene generated by inversion of chromosome 16 in human acute myeloid leukemia, is causative for oncogenic transformation. However, the mechanism by which CBFB-MYH11 initiates leukemogenesis is not clear. Previously published reports showed that CBFB-MYH11 dominantly inhibits RUNX1 and CBFB, and such inhibition has been suggested as the mechanism for leukemogenesis. Here we show that Cbfb-MYH11 caused Cbfb/Runx1 repression-independent defects in both primitive and definitive hematopoiesis. During primitive hematopoiesis, Cbfb-MYH11 delayed differentiation characterized by sustained expression of Gata2, Il1rl1, and Csf2rb, a phenotype not found in Cbfb and Runx1 knockout mice. Expression of Cbfb-MYH11 in the bone marrow induced the accumulation of abnormal progenitor-like cells expressing Csf2rb in preleukemic mice. The expression of all 3 genes was detected in most human and murine CBFB-MYH11(+) leukemia samples. Interestingly, Cbfb-MYH11(+) preleukemic progenitors and leukemia-initiating cells did not express Csf2rb, although the majority of leukemia cells in our Cbfb-MYH11 knockin mice were Csf2rb(+). Therefore Csf2rb can be used as a negative selection marker to enrich preleukemic progenitor cells and leukemia-initiating cells from Cbfb-MYH11 mice. These results suggest that Cbfb/Runx1 repression-independent activities contribute to leukemogenesis by Cbfb-MYH11.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Subunidade beta Comum dos Receptores de Citocinas/metabolismo , Células-Tronco Hematopoéticas/fisiologia , Leucemia Mieloide Aguda/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Animais , Apoptose/fisiologia , Biomarcadores , Diferenciação Celular/fisiologia , Divisão Celular/fisiologia , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade beta Comum dos Receptores de Citocinas/genética , Fator de Transcrição GATA2/genética , Fator de Transcrição GATA2/metabolismo , Regulação Leucêmica da Expressão Gênica , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/patologia , Humanos , Proteína 1 Semelhante a Receptor de Interleucina-1 , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas de Fusão Oncogênica/genética , Fenótipo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Regulação para Cima/fisiologia
12.
Nat Commun ; 13(1): 1982, 2022 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-35411048

RESUMO

T-cell large granular lymphocyte leukemia (T-LGLL) is a lymphoproliferative disease and bone marrow failure syndrome which responds to immunosuppressive therapies. We show single-cell TCR coupled with RNA sequencing of CD3+ T cells from 13 patients, sampled before and after alemtuzumab treatments. Effector memory T cells and loss of T cell receptor (TCR) repertoire diversity are prevalent in T-LGLL. Shared TCRA and TCRB clonotypes are absent. Deregulation of cell survival and apoptosis gene programs, and marked downregulation of apoptosis genes in CD8+ clones, are prominent features of T-LGLL cells. Apoptosis genes are upregulated after alemtuzumab treatment, especially in responders than non-responders; baseline expression levels of apoptosis genes are predictive of hematologic response. Alemtuzumab does not attenuate TCR clonality, and TCR diversity is further skewed after treatment. Inferences made from analysis of single cell data inform understanding of the pathophysiologic mechanisms of clonal expansion and persistence in T-LGLL.


Assuntos
Leucemia Linfocítica Granular Grande , Alemtuzumab/uso terapêutico , Células Clonais , Humanos , Leucemia Linfocítica Granular Grande/genética , Receptores de Antígenos de Linfócitos T/genética , Análise de Sequência de RNA
13.
J Leukoc Biol ; 111(2): 301-312, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34730257

RESUMO

Deficiency of adenosine deaminase 2 (DADA2) is a monogenic vasculitis syndrome caused by autosomal-recessive loss-of-function mutations in the ADA2 gene (previously known as CECR1). Vasculitis, vasculopathy, and inflammation are dominant clinical features of this disease; the spectrum of manifestations includes immunodeficiency and lymphoproliferation as well as hematologic manifestations. ADA2 is primarily secreted by stimulated monocytes and macrophages. Aberrant monocyte differentiation to macrophages and neutrophils are important in the pathogenesis of DADA2, but little is known about T lymphocytes in this disease. We performed combined single-cell RNA sequencing and single-cell TCR sequencing in order to profile T cell repertoires in 10 patients with DADA2. Although there were no significant alterations of T cell subsets, we observed activation of both CD8+ and CD4+ T cells. There was no clonal expansion of T cells: most TCRs were expressed at basal levels in patients and healthy donors. TCR usage was private to individual patients and not disease specific, indicating as unlikely a common pathogenic background or predisposition to a common pathogen. We recognized activation of IFN pathways as a signature of T cells and STAT1 as a hub gene in the gene network of T cell activation and cytotoxicity. Overall, T cells in DADA2 patients showed distinct cell-cell interactions with monocytes, as compared with healthy donors, and many of these ligand-receptor interactions likely drove up-regulation of STAT1 in both T cells and other immune cells in patients. Our analysis reveals previously undercharacterized cell characteristics in DADA2.


Assuntos
Adenosina Desaminase/deficiência , Biomarcadores/metabolismo , Regulação da Expressão Gênica , Síndromes de Imunodeficiência/patologia , Peptídeos e Proteínas de Sinalização Intercelular/deficiência , Dermatopatias/patologia , Linfócitos T/patologia , Doenças Vasculares/patologia , Adenosina Desaminase/genética , Adolescente , Adulto , Estudos de Casos e Controles , Células Cultivadas , Criança , Feminino , Seguimentos , Perfilação da Expressão Gênica , Humanos , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/imunologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Ativação Linfocitária , Masculino , Pessoa de Meia-Idade , Mutação , Prognóstico , Fator de Transcrição STAT1/genética , Análise de Célula Única , Dermatopatias/genética , Dermatopatias/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Doenças Vasculares/genética , Doenças Vasculares/imunologia , Adulto Jovem
14.
J Leukoc Biol ; 110(3): 409-424, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33988272

RESUMO

Deficiency of adenosine deaminase 2 (DADA2) is a rare autosomal recessive disease caused by loss-of-function variants in the ADA2 gene. DADA2 typically presents in childhood and is characterized by vasculopathy, stroke, inflammation, immunodeficiency, as well as hematologic manifestations. ADA2 protein is predominantly present in stimulated monocytes, dendritic cells, and macrophages. To elucidate molecular mechanisms in DADA2, CD14+ monocytes from 14 patients and 6 healthy donors were analyzed using single-cell RNA sequencing (scRNA-seq). Monocytes were purified by positive selection based on CD14 expression. Subpopulations were imputed from their transcriptomes. Based on scRNA-seq, monocytes could be classified as classical, intermediate, and nonclassical. Further, we used gene pathway analytics to interpret patterns of up- and down-regulated gene transcription. In DADA2, the frequency of nonclassical monocytes was higher compared with that of healthy donors, and M1 macrophage markers were up-regulated in patients. By comparing gene expression of each monocyte subtype between patients and healthy donors, we identified upregulated immune response pathways, including IFNα/ß and IFNγ signaling, in all monocyte subtypes. Distinctively, the TNFR2 noncanonical NF-κB pathway was up-regulated only in nonclassical monocytes. Patients' plasma showed increased IFNγ and TNFα levels. Our results suggest that elevated IFNγ activates cell signaling, leading to differentiation into M1 macrophages from monocytes and release of TNFα. Immune responses and more general response to stimuli pathways were up-regulated in DADA2 monocytes, and protein synthesis pathways were down-regulated, perhaps as stress responses. Our identification of novel aberrant immune pathways has implications for therapeutic approaches in DADA2 (registered at clinicaltrials.gov NCT00071045).


Assuntos
Agamaglobulinemia/genética , Agamaglobulinemia/patologia , Monócitos/patologia , Análise de Sequência de RNA , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/patologia , Análise de Célula Única , Adenosina Desaminase/genética , Adolescente , Adulto , Agamaglobulinemia/sangue , Agamaglobulinemia/enzimologia , Criança , Pré-Escolar , Citocinas/metabolismo , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Inflamação/patologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Interferons/metabolismo , Masculino , Pessoa de Meia-Idade , Mutação/genética , NF-kappa B/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Imunodeficiência Combinada Severa/sangue , Imunodeficiência Combinada Severa/enzimologia , Transdução de Sinais , Doadores de Tecidos , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA