Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
2.
Front Immunol ; 14: 1197490, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37398663

RESUMO

The Haematopoietically expressed homeobox transcription factor (Hhex) is a transcriptional repressor that is of fundamental importance across species, as evident by its evolutionary conservation spanning fish, amphibians, birds, mice and humans. Indeed, Hhex maintains its vital functions throughout the lifespan of the organism, beginning in the oocyte, through fundamental stages of embryogenesis in the foregut endoderm. The endodermal development driven by Hhex gives rise to endocrine organs such as the pancreas in a process which is likely linked to its role as a risk factor in diabetes and pancreatic disorders. Hhex is also required for the normal development of the bile duct and liver, the latter also importantly being the initial site of haematopoiesis. These haematopoietic origins are governed by Hhex, leading to its crucial later roles in definitive haematopoietic stem cell (HSC) self-renewal, lymphopoiesis and haematological malignancy. Hhex is also necessary for the developing forebrain and thyroid gland, with this reliance on Hhex evident in its role in endocrine disorders later in life including a potential role in Alzheimer's disease. Thus, the roles of Hhex in embryological development throughout evolution appear to be linked to its later roles in a variety of disease processes.


Assuntos
Genes Homeobox , Fatores de Transcrição , Humanos , Animais , Camundongos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Regulação da Expressão Gênica , Fígado/metabolismo , Sistema Digestório/metabolismo
3.
Nat Immunol ; 24(5): 814-826, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36997670

RESUMO

Missense mutations in PLCG2 can cause autoinflammation with phospholipase C gamma 2-associated antibody deficiency and immune dysregulation (APLAID). Here, we generated a mouse model carrying an APLAID mutation (p.Ser707Tyr) and found that inflammatory infiltrates in the skin and lungs were only partially ameliorated by removing inflammasome function via the deletion of caspase-1. Also, deleting interleukin-6 or tumor necrosis factor did not fully prevent APLAID mutant mice from autoinflammation. Overall, these findings are in accordance with the poor response individuals with APLAID have to treatments that block interleukin-1, JAK1/2 or tumor necrosis factor. Cytokine analysis revealed increased granulocyte colony-stimulating factor (G-CSF) levels as the most distinct feature in mice and individuals with APLAID. Remarkably, treatment with a G-CSF antibody completely reversed established disease in APLAID mice. Furthermore, excessive myelopoiesis was normalized and lymphocyte numbers rebounded. APLAID mice were also fully rescued by bone marrow transplantation from healthy donors, associated with reduced G-CSF production, predominantly from non-hematopoietic cells. In summary, we identify APLAID as a G-CSF-driven autoinflammatory disease, for which targeted therapy is feasible.


Assuntos
Transplante de Medula Óssea , Fator Estimulador de Colônias de Granulócitos , Animais , Camundongos , Citocinas , Interleucina-1 , Fator de Necrose Tumoral alfa/genética , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/imunologia , Síndromes de Imunodeficiência/metabolismo
4.
J Biol Chem ; 297(2): 100905, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34157287

RESUMO

Phosphatidylinositol-specific phospholipase Cγ2 (PLCγ2) is a critical signaling molecule activated downstream from a variety of cell surface receptors that contain an intracellular immunoreceptor tyrosine-based activation motif. These receptors recruit kinases such as Syk, BTK, and BLNK to phosphorylate and activate PLCγ2, which then generates 1D-myo-inositol 1,4,5-trisphosphate and diacylglycerol. These well-known second messengers are required for diverse membrane functionality including cellular proliferation, endocytosis, and calcium flux. As a result, PLCγ2 dysfunction is associated with a variety of diseases including cancer, neurodegeneration, and immune disorders. The diverse pathologies associated with PLCγ2 are exemplified by distinct genetic variants. Inherited mutations at this locus cause PLCγ2-associated antibody deficiency and immune dysregulation, in some cases with autoinflammation. Acquired mutations at this locus, which often arise as a result of BTK inhibition to treat chronic lymphocytic leukemia, result in constitutive downstream signaling and lymphocyte proliferation. Finally, a third group of PLCγ2 variants actually has a protective effect in a variety of neurodegenerative disorders, presumably by increased uptake and degradation of deleterious neurological aggregates. Therefore, manipulating PLCγ2 activity either up or down could have therapeutic benefit; however, we require a better understanding of the signaling pathways propagated by these variants before such clinical utility can be realized. Here, we review the signaling roles of PLCγ2 in hematopoietic cells to help understand the effect of mutations driving immune disorders and cancer and extrapolate from this to roles which may relate to protection against neurodegeneration.


Assuntos
Tirosina Quinase da Agamaglobulinemia/imunologia , Sinalização do Cálcio , Doenças do Sistema Imunitário/patologia , Neoplasias/patologia , Doenças Neurodegenerativas/patologia , Fosfolipase C gama/metabolismo , Quinase Syk/imunologia , Tirosina Quinase da Agamaglobulinemia/metabolismo , Animais , Humanos , Doenças do Sistema Imunitário/genética , Doenças do Sistema Imunitário/imunologia , Doenças do Sistema Imunitário/metabolismo , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/metabolismo , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/imunologia , Doenças Neurodegenerativas/metabolismo , Fosfolipase C gama/genética , Fosfolipase C gama/imunologia , Quinase Syk/metabolismo
5.
Blood ; 137(21): 2920-2934, 2021 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-33512466

RESUMO

OBF1 is a specific coactivator of the POU family transcription factors OCT1 and OCT2. OBF1 and OCT2 are B cell-specific and indispensable for germinal center (GC) formation, but their mechanism of action is unclear. Here, we show by chromatin immunoprecipitation-sequencing that OBF1 extensively colocalizes with OCT1 and OCT2. We found that these factors also often colocalize with transcription factors of the ETS family. Furthermore, we showed that OBF1, OCT2, and OCT1 bind widely to the promoters or enhancers of genes involved in GC formation in mouse and human GC B cells. Short hairpin RNA knockdown experiments demonstrated that OCT1, OCT2, and OBF1 regulate each other and are essential for proliferation of GC-derived lymphoma cell lines. OBF1 downregulation disrupts the GC transcriptional program: genes involved in GC maintenance, such as BCL6, are downregulated, whereas genes related to exit from the GC program, such as IRF4, are upregulated. Ectopic expression of BCL6 does not restore the proliferation of GC-derived lymphoma cells depleted of OBF1 unless IRF4 is also depleted, indicating that OBF1 controls an essential regulatory node in GC differentiation.


Assuntos
Centro Germinativo/metabolismo , Fator 1 de Transcrição de Octâmero/fisiologia , Fator 2 de Transcrição de Octâmero/uso terapêutico , Transativadores/uso terapêutico , Transcrição Gênica/genética , Animais , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Ontologia Genética , Células HEK293 , Humanos , Lipopolissacarídeos/farmacologia , Linfoma não Hodgkin/genética , Linfoma não Hodgkin/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fator 1 de Transcrição de Octâmero/deficiência , Fator 1 de Transcrição de Octâmero/genética , Fator 2 de Transcrição de Octâmero/deficiência , Fator 2 de Transcrição de Octâmero/genética , Proteína Proto-Oncogênica c-ets-1/análise , Interferência de RNA , RNA Interferente Pequeno/genética , Proteínas Recombinantes/metabolismo , Transativadores/deficiência , Transativadores/genética
6.
Leukemia ; 35(8): 2205-2219, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33483615

RESUMO

The majority of cases of T-cell acute lymphoblastic leukemia (T-ALL) contain chromosomal abnormalities that drive overexpression of oncogenic transcription factors. However, whether these initiating oncogenes are required for leukemia maintenance is poorly understood. To address this, we developed a tetracycline-regulated mouse model of T-ALL driven by the oncogenic transcription factor Lmo2. This revealed that whilst thymus-resident pre-Leukemic Stem Cells (pre-LSCs) required continuous Lmo2 expression, the majority of leukemias relapsed despite Lmo2 withdrawal. Relapse was associated with a mature phenotype and frequent mutation or loss of tumor suppressor genes including Ikzf1 (Ikaros), with targeted deletion Ikzf1 being sufficient to transform Lmo2-dependent leukemias to Lmo2-independence. Moreover, we found that the related transcription factor TAL1 was dispensable in several human T-ALL cell lines that contain SIL-TAL1 chromosomal deletions driving its overexpression, indicating that evolution to oncogene independence can also occur in human T-ALL. Together these results indicate an evolution of oncogene addiction in murine and human T-ALL and show that loss of Ikaros is a mechanism that can promote self-renewal of T-ALL lymphoblasts in the absence of an initiating oncogenic transcription factor.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Regulação Leucêmica da Expressão Gênica , Fator de Transcrição Ikaros/fisiologia , Proteínas com Domínio LIM/fisiologia , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patologia , Animais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oncogenes , Leucemia-Linfoma Linfoblástico de Células T Precursoras/etiologia , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo
8.
Nat Immunol ; 21(12): 1574-1584, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33077975

RESUMO

A classical view of blood cell development is that multipotent hematopoietic stem and progenitor cells (HSPCs) become lineage-restricted at defined stages. Lin-c-Kit+Sca-1+Flt3+ cells, termed lymphoid-primed multipotent progenitors (LMPPs), have lost megakaryocyte and erythroid potential but are heterogeneous in their fate. Here, through single-cell RNA sequencing, we identify the expression of Dach1 and associated genes in this fraction as being coexpressed with myeloid/stem genes but inversely correlated with lymphoid genes. Through generation of Dach1-GFP reporter mice, we identify a transcriptionally and functionally unique Dach1-GFP- subpopulation within LMPPs with lymphoid potential with low to negligible classic myeloid potential. We term these 'lymphoid-primed progenitors' (LPPs). These findings define an early definitive branch point of lymphoid development in hematopoiesis and a means for prospective isolation of LPPs.


Assuntos
Biomarcadores , Proteínas do Olho/metabolismo , Genômica , Células Progenitoras Linfoides/metabolismo , Análise de Célula Única , Animais , Células Cultivadas , Biologia Computacional/métodos , Proteínas do Olho/genética , Perfilação da Expressão Gênica , Genômica/métodos , Hematopoese/genética , Sequenciamento de Nucleotídeos em Larga Escala , Células Progenitoras Linfoides/citologia , Células Progenitoras Linfoides/imunologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteômica , Análise de Célula Única/métodos
9.
Eur J Immunol ; 50(7): 959-971, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32090320

RESUMO

The transcription factor Hhex (hematopoietically expressed homeobox gene) is critical for development of multiple lymphoid lineages beyond the common lymphoid progenitor. In addition, Hhex regulates hematopoietic stem cell (HSC) self-renewal, emergency hematopoiesis, and acute myeloid leukemia initiation and maintenance. Hhex mediates its effects on HSCs and acute myeloid leukemia stem cells via repression of the Cdkn2a tumor suppressor locus. However, we report here that loss of Cdkn2a does not rescue the failure of lymphoid development caused by loss of Hhex. As loss of Hhex causes apoptosis of lymphoid progenitors associated with impaired Bcl2 expression and defective Stat5b signaling, we tested the effects of rescuing these pathways using transgenic mice. Expression of the anti-apoptotic factor Bcl2, but not activated Stat5, rescued the development of T-, B-, and NK-cell lineages in the absence of Hhex. These results indicate that Bcl2 expression, but not Stat5b signaling or loss of Cdkn2a, can overcome the lymphoid deficiencies caused by the absence of Hhex, suggesting that the primary role of this transcription factor is to promote survival of lymphoid progenitors during early lymphoid development.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/imunologia , Proteínas de Homeodomínio/imunologia , Células Progenitoras Linfoides/imunologia , Fator de Transcrição STAT5/imunologia , Transdução de Sinais/imunologia , Fatores de Transcrição/imunologia , Animais , Apoptose/genética , Apoptose/imunologia , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Inibidor p16 de Quinase Dependente de Ciclina/genética , Proteínas de Homeodomínio/genética , Células Progenitoras Linfoides/citologia , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/imunologia , Fator de Transcrição STAT5/genética , Transdução de Sinais/genética , Fatores de Transcrição/genética
10.
Leukemia ; 33(8): 1868-1880, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30700838

RESUMO

T cell acute lymphoblastic leukaemia (T-ALL) cases include subfamilies that overexpress the TAL1/LMO, TLX1/3 and HOXA transcription factor oncogenes. While it has been shown that TAL1/LMO transcription factors induce self-renewal of thymocytes, whether this is true for other transcription factor oncogenes is unknown. To address this, we have studied NUP98-HOXD13-transgenic (NHD13-Tg) mice, which overexpress HOXA transcription factors throughout haematopoiesis and develop both myelodysplastic syndrome (MDS) progressing to acute myeloid leukaemia (AML) as well as T-ALL. We find that thymocytes from preleukaemic NHD13-Tg mice can serially transplant, demonstrating that they have self-renewal capacity. Transcriptome analysis shows that NHD13-Tg thymocytes exhibit a stem cell-like transcriptional programme closely resembling that induced by Lmo2, including Lmo2 itself and its critical cofactor Lyl1. To determine whether Lmo2/Lyl1 are required for NHD13-induced thymocyte self-renewal, NHD13-Tg mice were crossed with Lyl1 knockout mice. This showed that Lyl1 is essential for expression of the stem cell-like gene expression programme in thymocytes and self-renewal. Surprisingly however, NHD13 transgenic mice lacking Lyl1 showed accelerated T-ALL and absence of transformation to AML, associated with a loss of multipotent progenitors in the bone marrow. Thus multiple T cell oncogenes induce thymocyte self-renewal via Lmo2/Lyl1; however, NHD13 can also promote T-ALL via an alternative pathway.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Proteínas de Homeodomínio/genética , Proteínas com Domínio LIM/fisiologia , Proteínas de Neoplasias/fisiologia , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Proteínas de Fusão Oncogênica/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Timócitos/fisiologia , Fatores de Transcrição/genética , Animais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
11.
Blood Adv ; 2(4): 347-360, 2018 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-29453249

RESUMO

The hematopoietically expressed homeobox (Hhex) transcription factor is overexpressed in human myeloid leukemias. Conditional knockout models of murine acute myeloid leukemia indicate that Hhex maintains leukemia stem cell self-renewal by enabling Polycomb-mediated epigenetic repression of the Cdkn2a tumor suppressor locus, encoding p16Ink4a and p19Arf However, whether Hhex overexpression also affects hematopoietic differentiation is unknown. To study this, we retrovirally overexpressed Hhex in hematopoietic progenitors. This enabled serial replating of myeloid progenitors, leading to the rapid establishment of interleukin-3 (IL-3)-dependent promyelocytic cell lines. Use of a Hhex-ERT2 fusion protein demonstrated that continuous nuclear Hhex is required for transformation, and structure function analysis demonstrated a requirement of the DNA-binding and N-terminal-repressive domains of Hhex for promyelocytic transformation. This included the N-terminal promyelocytic leukemia protein (Pml) interaction domain, although deletion of Pml failed to prevent Hhex-induced promyelocyte transformation, implying other critical partners. Furthermore, deletion of p16Ink4a or p19Arf did not promote promyelocyte transformation, indicating that repression of distinct Hhex target genes is required for this process. Indeed, transcriptome analysis showed that Hhex overexpression resulted in repression of several myeloid developmental genes. To test the potential for Hhex overexpression to contribute to leukemic transformation, Hhex-transformed promyelocyte lines were rendered growth factor-independent using a constitutively active IL-3 receptor common ß subunit (ßcV449E). The resultant cell lines resulted in a rapid promyelocytic leukemia in vivo. Thus, Hhex overexpression can contribute to myeloid leukemia via multiple mechanisms including differentiation blockade and enabling epigenetic repression of the Cdkn2a locus.


Assuntos
Autorrenovação Celular , Células Precursoras de Granulócitos/citologia , Proteínas de Homeodomínio/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular , Leucemia Mieloide/etiologia , Fatores de Transcrição/fisiologia , Animais , Técnicas de Cultura de Células , Diferenciação Celular , Núcleo Celular/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Regulação Leucêmica da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Camundongos , Fatores de Transcrição/metabolismo
12.
Stem Cells ; 35(8): 1948-1957, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28577303

RESUMO

The hematopoietically expressed homeobox transcription factor (Hhex) is important for the maturation of definitive hematopoietic progenitors and B-cells during development. We have recently shown that in adult hematopoiesis, Hhex is dispensable for maintenance of hematopoietic stem cells (HSCs) and myeloid lineages but essential for the commitment of common lymphoid progenitors (CLPs) to lymphoid lineages. Here, we show that during serial bone marrow transplantation, Hhex-deleted HSCs are progressively lost, revealing an intrinsic defect in HSC self-renewal. Moreover, Hhex-deleted mice show markedly impaired hematopoietic recovery following myeloablation, due to a failure of progenitor expansion. In vitro, Hhex-null blast colonies were incapable of replating, implying a specific requirement for Hhex in immature progenitors. Transcriptome analysis of Hhex-null Lin- Sca+ Kit+ cells showed that Hhex deletion leads to derepression of polycomb repressive complex 2 (PRC2) and PRC1 target genes, including the Cdkn2a locus encoding the tumor suppressors p16Ink 4a and p19Arf . Indeed, loss of Cdkn2a restored the capacity of Hhex-null blast colonies to generate myeloid progenitors in vitro, as well as hematopoietic reconstitution following myeloablation in vivo. Thus, HSCs require Hhex to promote PRC2-mediated Cdkn2a repression to enable continued self-renewal and response to hematopoietic stress. Stem Cells 2017;35:1948-1957.


Assuntos
Autorrenovação Celular , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Hematopoese , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Proteínas de Homeodomínio/metabolismo , Estresse Fisiológico , Fatores de Transcrição/metabolismo , Animais , Proliferação de Células , Deleção de Genes , Regulação da Expressão Gênica , Transplante de Células-Tronco Hematopoéticas , Camundongos Endogâmicos C57BL , Células Progenitoras Mieloides/citologia , Células Progenitoras Mieloides/metabolismo
13.
Genes Dev ; 30(1): 78-91, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26728554

RESUMO

Unlike clustered HOX genes, the role of nonclustered homeobox gene family members in hematopoiesis and leukemogenesis has not been extensively studied. Here we found that the hematopoietically expressed homeobox gene Hhex is overexpressed in acute myeloid leukemia (AML) and is essential for the initiation and propagation of MLL-ENL-induced AML but dispensable for normal myelopoiesis, indicating a specific requirement for Hhex for leukemic growth. Loss of Hhex leads to expression of the Cdkn2a-encoded tumor suppressors p16(INK4a) and p19(ARF), which are required for growth arrest and myeloid differentiation following Hhex deletion. Mechanistically, we show that Hhex binds to the Cdkn2a locus and directly interacts with the Polycomb-repressive complex 2 (PRC2) to enable H3K27me3-mediated epigenetic repression. Thus, Hhex is a potential therapeutic target that is specifically required for AML stem cells to repress tumor suppressor pathways and enable continued self-renewal.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/genética , Epigênese Genética , Proteínas de Homeodomínio/metabolismo , Leucemia Mieloide Aguda/fisiopatologia , Complexo Repressor Polycomb 2/genética , Complexo Repressor Polycomb 2/metabolismo , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular Tumoral , Deleção de Genes , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/genética , Humanos , Leucemia Mieloide Aguda/genética , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica , Fatores de Transcrição/genética
14.
Blood ; 125(5): 803-14, 2015 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-25472970

RESUMO

The hematopoietically expressed homeobox gene, Hhex, is a transcription factor that is important for development of definitive hematopoietic stem cells (HSCs) and B cells, and that causes T-cell leukemia when overexpressed. Here, we have used an Hhex inducible knockout mouse model to study the role of Hhex in adult hematopoiesis. We found that loss of Hhex was tolerated in HSCs and myeloid lineages, but resulted in a progressive loss of B lymphocytes in the circulation. This was accompanied by a complete loss of B-cell progenitors in the bone marrow and of transitional B-cell subsets in the spleen. In addition, transplantation and in vitro culture experiments demonstrated an almost complete failure of Hhex-null HSCs to contribute to lymphoid lineages beyond the common lymphoid precursor stage, including T cells, B cells, NK cells, and dendritic cells. Gene expression analysis of Hhex-deleted progenitors demonstrated deregulated expression of a number of cell cycle regulators. Overexpression of one of these, cyclin D1, could rescue the B-cell developmental potential of Hhex-null lymphoid precursors. Thus, Hhex is a key regulator of early lymphoid development, functioning, at least in part, via regulation of the cell cycle.


Assuntos
Proteínas de Ciclo Celular/genética , Ciclina D1/genética , Hematopoese/genética , Proteínas de Homeodomínio/genética , Linfopoese/genética , Células Precursoras de Linfócitos B/patologia , Fatores de Transcrição/genética , Animais , Células da Medula Óssea/imunologia , Células da Medula Óssea/patologia , Proteínas de Ciclo Celular/imunologia , Diferenciação Celular , Proliferação de Células , Ciclina D1/imunologia , Células Dendríticas/imunologia , Células Dendríticas/patologia , Deleção de Genes , Regulação da Expressão Gênica , Teste de Complementação Genética , Hematopoese/imunologia , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/patologia , Proteínas de Homeodomínio/imunologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/patologia , Contagem de Linfócitos , Depleção Linfocítica , Linfopoese/imunologia , Camundongos , Camundongos Knockout , Células Precursoras de Linfócitos B/imunologia , Baço/imunologia , Baço/patologia , Linfócitos T/imunologia , Linfócitos T/patologia , Fatores de Transcrição/deficiência , Fatores de Transcrição/imunologia , Transcrição Gênica
15.
Blood ; 122(12): 2093-103, 2013 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-23926305

RESUMO

Lmo2 is an oncogenic transcription factor that is frequently overexpressed in T-cell acute lymphoblastic leukemia (T-ALL), including early T-cell precursor ALL (ETP-ALL) cases with poor prognosis. Lmo2 must be recruited to DNA by binding to the hematopoietic basic helix-loop-helix factors Scl/Tal1 or Lyl1. However, it is unknown which of these factors can mediate the leukemic activity of Lmo2. To address this, we have generated Lmo2-transgenic mice lacking either Scl or Lyl1 in the thymus. We show that although Scl is dispensable for Lmo2-driven leukemia, Lyl1 is critical for all oncogenic functions of Lmo2, including upregulation of a stem cell-like gene signature, aberrant self-renewal of thymocytes, and subsequent generation of T-cell leukemia. Lyl1 expression is restricted to preleukemic and leukemic stem cell populations in this model, providing a molecular explanation for the stage-specific expression of the Lmo2-induced gene expression program. Moreover, LMO2 and LYL1 are coexpressed in ETP-ALL patient samples, and LYL1 is required for growth of ETP-ALL cell lines. Thus, the LMO2-LYL1 interaction is a promising therapeutic target for inhibiting self-renewing cancer stem cells in T-ALL, including poor-prognosis ETP-ALL cases.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Proteínas com Domínio LIM/genética , Proteínas de Neoplasias/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular/genética , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Análise por Conglomerados , Perfilação da Expressão Gênica , Regulação Leucêmica da Expressão Gênica , Humanos , Proteínas com Domínio LIM/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas de Neoplasias/metabolismo , Células-Tronco Neoplásicas/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/mortalidade , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteína 1 de Leucemia Linfocítica Aguda de Células T , Linfócitos T/citologia , Linfócitos T/metabolismo , Timócitos/metabolismo , Timócitos/patologia
16.
Phys Biol ; 8(6): 066002, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21946082

RESUMO

The epidermal growth factor receptor (EGFR) is a member of the erbB tyrosine kinase family of receptors. Structural studies have revealed two distinct conformations of the ectodomain of the EGFR: a compact, tethered, conformation and an untethered extended conformation. In the context of a monomer-dimer transition model, ligand binding is thought to untether the monomeric receptor leading to exposure of a dimerization arm which then facilitates receptor dimerization, kinase activation and signaling. For receptors directed orthogonal to the local plane of the membrane surface, this would lead to a large change in the distance of the receptor N-terminus from the membrane surface. To investigate this experimentally, we produced stable BaF/3 cell lines expressing a biochemically functional yellow fluorescent protein (YFP)-EGFR chimera and determined the vertical separation of the N-terminal YFP tag from the membrane using fluorescence resonance energy transfer (FRET) techniques. Homo-FRET/rFLIM was employed to determine the presence of unliganded dimers and to measure the average distance between the N-terminal tags in those dimers. The results suggest that EGF-induced activation occurs within or between pre-formed and extended dimers with very little change in the extension of the N-terminii from the membrane surface. These results provide constraints on possible models for EGFR activation.


Assuntos
Proteínas de Bactérias/química , Receptores ErbB/química , Proteínas Luminescentes/química , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Linhagem Celular , Receptores ErbB/genética , Receptores ErbB/metabolismo , Transferência Ressonante de Energia de Fluorescência , Expressão Gênica , Ligantes , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Multimerização Proteica , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
17.
J Immunol ; 181(5): 3449-55, 2008 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-18714017

RESUMO

NK cells hold great potential for improving the immunotherapy of cancer. Nevertheless, tumor cells can effectively escape NK cell-mediated apoptosis through interaction of MHC molecules with NK cell inhibitory receptors. Thus, to harness NK cell effector function against tumors, we used Amaxa gene transfer technology to gene-modify primary mouse NK cells with a chimeric single-chain variable fragment (scFv) receptor specific for the human erbB2 tumor-associated Ag. The chimeric receptor was composed of the extracellular scFv anti-erbB2 Ab linked to the transmembrane and cytoplasmic CD28 and TCR-zeta signaling domains (scFv-CD28-zeta). In this study we demonstrated that mouse NK cells gene-modified with this chimera could specifically mediate enhanced killing of an erbB2(+) MHC class I(+) lymphoma in a perforin-dependent manner. Expression of the chimera did not interfere with NK cell-mediated cytotoxicity mediated by endogenous NK receptors. Furthermore, adoptive transfer of gene-modified NK cells significantly enhanced the survival of RAG mice bearing established i.p. RMA-erbB2(+) lymphoma. In summary, these data suggest that use of genetically modified NK cells could broaden the scope of cancer immunotherapy for patients.


Assuntos
Imunoterapia Adotiva/métodos , Células Matadoras Naturais/transplante , Linfoma/terapia , Proteínas Recombinantes de Fusão/uso terapêutico , Animais , Anticorpos/genética , Antígenos CD28/genética , Citotoxicidade Imunológica , Progressão da Doença , Técnicas de Transferência de Genes , Humanos , Região Variável de Imunoglobulina , Células Matadoras Naturais/metabolismo , Linfoma/mortalidade , Camundongos , Camundongos Endogâmicos C57BL , Receptor ErbB-2/imunologia , Receptores de Antígenos de Linfócitos T/genética , Proteínas Recombinantes de Fusão/genética , Taxa de Sobrevida
18.
Hum Gene Ther ; 17(11): 1134-43, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17052145

RESUMO

Immunotherapeutic approaches involving genetic modification of T cells show promise in generating highly specific tumor-reactive effector cells for cancer treatment. Given the high affinity of FcRI (the subtype I Fc receptor for IgE) for IgE monoclonal antibody (mAb), modification of T cells with chimeric FcRI in combination with tumor-specific IgE mAbs is potentially a powerful and effective strategy to specifically target T cells to tumor cells. In this study, we retrovirally transduce human primary T cells with a cDNA encoding the extracellular domain of FcRI linked to the hinge and transmembrane domains of FcRI and the cytoplasmic domains of CD28 and T cell receptor zeta chain (FcRI-CD28-zeta). We demonstrate that human T cells expressing FcRI-CD28-zeta, in the presence of tumor-specific IgE mAb recognizing mouse CD8 antigen (Ly- 2.1+), can specifically secrete cytokine, proliferate, and mediate cytotoxic function after antigen ligation. Furthermore, adoptive transfer of FcRI-CD28-zeta cells incubated with anti-Ly-2.1 IgE mAb significantly enhances the survival of irradiated nonobese diabetic-severe combined immunodeficiency mice bearing Ly-2.1+ tumor compared with control mice. Thus, this set of experiments demonstrates that Fc gene-engineered human T cells mediate effector function in vitro and in vivo in an IgE-dependent manner and thus a novel and valid approach for cancer therapy can now be further developed.


Assuntos
Transferência Adotiva , Vacinas Anticâncer , Imunoterapia , Receptores de IgE/genética , Receptores de IgE/uso terapêutico , Linfócitos T/transplante , Timoma/terapia , Animais , Sequência de Bases , Antígenos CD28/genética , Linhagem Celular , Proliferação de Células , Citocinas/metabolismo , Testes Imunológicos de Citotoxicidade , Modelos Animais de Doenças , Humanos , Imunoglobulina E/sangue , Proteínas de Membrana/genética , Camundongos , Dados de Sequência Molecular , Receptores de Antígenos de Linfócitos T/genética , Receptores de IgE/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/uso terapêutico , Taxa de Sobrevida , Linfócitos T/imunologia , Timoma/imunologia , Transdução Genética
19.
Blood ; 106(9): 2995-3003, 2005 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-16030195

RESUMO

Because CD4+ T cells play a key role in aiding cellular immune responses, we wanted to assess whether increasing numbers of gene-engineered antigen-restricted CD4+ T cells could enhance an antitumor response mediated by similarly gene-engineered CD8+ T cells. In this study, we have used retroviral transduction to generate erbB2-reactive mouse T-cell populations composed of various proportions of CD4+ and CD8+ cells and then determined the antitumor reactivity of these mixtures. Gene-modified CD4+ and CD8+ T cells were shown to specifically secrete Tc1 (T cytotoxic-1) or Tc2 cytokines, proliferate, and lyse erbB2+ tumor targets following antigen ligation in vitro. In adoptive transfer experiments using severe combined immunodeficient (scid) mice, we demonstrated that injection of equivalent numbers of antigen-specific engineered CD8+ and CD4+ T cells led to significant improvement in survival of mice bearing established lung metastases compared with transfer of unfractionated (largely CD8+) engineered T cells. Transferred CD4+ T cells had to be antigen-specific (not just activated) and secrete interferon gamma (IFN-gamma) to potentiate the antitumor effect. Importantly, antitumor responses in these mice correlated with localization and persistence of gene-engineered T cells at the tumor site. Strikingly, mice that survived primary tumor challenge could reject a subsequent rechallenge. Overall, this study has highlighted the therapeutic potential of using combined transfer of antigen-specific gene-modified CD8+ and CD4+ T cells to significantly enhance T-cell adoptive transfer strategies for cancer therapy.


Assuntos
Transferência Adotiva , Engenharia Genética , Neoplasias/imunologia , Neoplasias/terapia , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo , Animais , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Citocinas/metabolismo , Regulação da Expressão Gênica , Humanos , Interferon gama/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Neoplasias/metabolismo , Neoplasias/patologia , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Anticorpos de Cadeia Única , Taxa de Sobrevida
20.
Biochem Biophys Res Commun ; 326(2): 417-24, 2005 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-15582594

RESUMO

The human HIN-200 family member AIM2 was originally identified in a screen for suppressors of melanoma tumorigenicity following introduction of chromosome 6 into the UACC903 human melanoma cell line. Although the AIM2 protein contained many of the conserved structural motifs common to other HIN-200 proteins, the biochemical characteristics of AIM2 and the ability of overexpressed AIM2 to phenocopy the effect of introduction of chromosome 6 in the UACC903 cells had not been assessed. Herein we demonstrated that AIM2 was localised within the nucleus of transfected or interferon-treated human cells. In addition, AIM2 could homodimerise via the amino-terminal (PAAD/DAPIN) region and heterodimerise with the related IFI 16 protein. However, overexpressed AIM2 did not significantly affect the growth or survival of UACC903 cells or another human melanoma cell line. These data indicate that AIM2 has many of the biochemical and structural characteristics of HIN-200 proteins, however, its expression is not sufficient to induce a tumor-suppressor-like phenotype.


Assuntos
Família Multigênica , Proteínas Nucleares/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Anticorpos Monoclonais/imunologia , Linhagem Celular , Proliferação de Células , Proteínas de Ligação a DNA , Dimerização , Expressão Gênica , Humanos , Interferons/farmacologia , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/imunologia , Estrutura Quaternária de Proteína , Transporte Proteico , Proteínas Supressoras de Tumor/química , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA