Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
JCI Insight ; 8(19)2023 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-37643018

RESUMO

The role of aberrant glycosylation in pancreatic ductal adenocarcinoma (PDAC) remains an under-investigated area of research. In this study, we determined that ST6 ß-galactoside α2,6 sialyltransferase 1 (ST6GAL1), which adds α2,6-linked sialic acids to N-glycosylated proteins, was upregulated in patients with early-stage PDAC and was further increased in advanced disease. A tumor-promoting function for ST6GAL1 was elucidated using tumor xenograft experiments with human PDAC cells. Additionally, we developed a genetically engineered mouse (GEM) model with transgenic expression of ST6GAL1 in the pancreas and found that mice with dual expression of ST6GAL1 and oncogenic KRASG12D had greatly accelerated PDAC progression compared with mice expressing KRASG12D alone. As ST6GAL1 imparts progenitor-like characteristics, we interrogated ST6GAL1's role in acinar to ductal metaplasia (ADM), a process that fosters neoplasia by reprogramming acinar cells into ductal, progenitor-like cells. We verified ST6GAL1 promotes ADM using multiple models including the 266-6 cell line, GEM-derived organoids and tissues, and an in vivo model of inflammation-induced ADM. EGFR is a key driver of ADM and is known to be activated by ST6GAL1-mediated sialylation. Importantly, EGFR activation was dramatically increased in acinar cells and organoids from mice with transgenic ST6GAL1 expression. These collective results highlight a glycosylation-dependent mechanism involved in early stages of pancreatic neoplasia.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Camundongos , Animais , Neoplasias Pancreáticas/patologia , Pâncreas/patologia , Carcinoma Ductal Pancreático/patologia , Receptores ErbB/genética , Metaplasia/patologia , Sialiltransferases/genética , beta-D-Galactosídeo alfa 2-6-Sialiltransferase , Antígenos CD
2.
Cell Rep ; 36(4): 109421, 2021 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-34320342

RESUMO

Mitogen-activated protein kinases (MAPKs) are inactivated by dual-specificity phosphatases (DUSPs), the activities of which are tightly regulated during cell differentiation. Using knockdown screening and single-cell transcriptional analysis, we demonstrate that DUSP4 is the phosphatase that specifically inactivates p38 kinase to promote megakaryocyte (Mk) differentiation. Mechanistically, PRMT1-mediated methylation of DUSP4 triggers its ubiquitinylation by an E3 ligase HUWE1. Interestingly, the mechanistic axis of the DUSP4 degradation and p38 activation is also associated with a transcriptional signature of immune activation in Mk cells. In the context of thrombocytopenia observed in myelodysplastic syndrome (MDS), we demonstrate that high levels of p38 MAPK and PRMT1 are associated with low platelet counts and adverse prognosis, while pharmacological inhibition of p38 MAPK or PRMT1 stimulates megakaryopoiesis. These findings provide mechanistic insights into the role of the PRMT1-DUSP4-p38 axis on Mk differentiation and present a strategy for treatment of thrombocytopenia associated with MDS.


Assuntos
Diferenciação Celular , Fosfatases de Especificidade Dupla , Megacariócitos , Fosfatases da Proteína Quinase Ativada por Mitógeno , Adulto , Animais , Criança , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Adulto Jovem , Arginina/metabolismo , Linhagem Celular , Fosfatases de Especificidade Dupla/metabolismo , Estabilidade Enzimática , Células HEK293 , Sistema de Sinalização das MAP Quinases , Megacariócitos/citologia , Megacariócitos/enzimologia , Metilação , Camundongos Endogâmicos C57BL , Fosfatases da Proteína Quinase Ativada por Mitógeno/metabolismo , Síndromes Mielodisplásicas/enzimologia , Síndromes Mielodisplásicas/patologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Poliubiquitina/metabolismo , Proteína-Arginina N-Metiltransferases/antagonistas & inibidores , Proteína-Arginina N-Metiltransferases/metabolismo , Proteólise , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/metabolismo , Ubiquitinação
3.
Blood Adv ; 2(21): 2829-2836, 2018 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-30373889

RESUMO

Dynamic regulation of histone modification enzymes such as PRMT1 (protein arginine methyltransferase 1) determines the ordered epigenetic transitions in hematopoiesis. Sorting cells according to the expression levels of histone modification enzymes may further define subpopulations in hematopoietic lineages with unique differentiation potentials that are presently defined by surface markers. We discovered a vital near infrared dye, E84, that fluoresces brightly following binding to PRMT1 and excitation with a red laser. The staining intensity as measured by flow cytometry is correlated with the PRMT1 expression level. Importantly, E84 staining has no apparent negative effect on the proliferation of the labeled cells. Given that long-term hematopoietic stem cells (LT-HSCs) produce low levels of PRMT1, we used E84 to sort LT-HSCs from mouse bone marrow. We found that SLAM (the signalling lymphocyte activation molecule family) marker-positive LT-HSCs were enriched in the E84low cell fraction. We then performed bone marrow transplantations with E84high or E84low Lin-Sca1+Kit+ (LSK) cells and showed that whole blood cell lineages were successfully reconstituted 16 weeks after transplanting 200 E84low LSK cells. Thus, E84 is a useful new tool to probe the role of PRMT1 in hematopoiesis and leukemogenesis. Developing E84 and other small molecules to label histone modification enzymes provides a convenient approach without modifying gene loci to study the interaction between hematopoietic stem/progenitor cell epigenetic status and differentiation state.


Assuntos
Células Sanguíneas/metabolismo , Carbocianinas/química , Epigênese Genética , Corantes Fluorescentes/química , Proteína-Arginina N-Metiltransferases/genética , Animais , Ataxina-1/metabolismo , Células Sanguíneas/patologia , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Transplante de Medula Óssea , Linhagem da Célula , Citometria de Fluxo/métodos , Células HEK293 , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Humanos , Antígenos Comuns de Leucócito/metabolismo , Camundongos , Proteína-Arginina N-Metiltransferases/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo
4.
Stem Cells ; 35(2): 473-484, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27539014

RESUMO

Long-term self-renewing hematopoietic stem cell (LT-HSC) homeostasis within the bone marrow (BM) of adult mammals is regulated by complex interactions between LT-HSC and a number of niche-associated cell types including mesenchymal stromal/stem cells (MSC), osteoblasts (OB), macrophage, and neuronal cells in close proximity with the vasculature. Here, we cloned and functionally characterized a murine BM MSC subpopulation that was uniformly Nestin+ Lepr + Sca-1+ CD146+ and could be stably propagated with high colony-forming unit fibroblast re-cloning efficiency. MSC synergized with SCF and IL-11 to support a 20-fold expansion in true LT-HSC after 10-days of in vitro coculture. Optimal stimulation of LT-HSC expansion was minimally dependent on Notch signaling but was significantly enhanced by global inhibition of Wnt signaling. The self-renewal-promoting activity of MSC was progressively lost when MSC clones were differentiated into mature OB. This suggests that the stage of osteoblast development may significantly impact the ability of osteolineage cells to support LT-HSC homeostasis in vivo. Stem Cells 2017;35:473-484.


Assuntos
Autorrenovação Celular , Células-Tronco Hematopoéticas/citologia , Células-Tronco Mesenquimais/citologia , Osteogênese , Animais , Células da Medula Óssea/citologia , Diferenciação Celular , Células Cultivadas , Células Clonais , Técnicas de Cocultura , Células-Tronco Hematopoéticas/metabolismo , Camundongos Endogâmicos C57BL , Ossificação Heterotópica/patologia , Receptores Notch/metabolismo , Transdução de Sinais
5.
Cancer Res ; 76(13): 3978-88, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27216178

RESUMO

The glycosyltransferase ST6Gal-I, which adds α2-6-linked sialic acids to substrate glycoproteins, has been implicated in carcinogenesis; however, the nature of its pathogenic role remains poorly understood. Here we show that ST6Gal-I is upregulated in ovarian and pancreatic carcinomas, enriched in metastatic tumors, and associated with reduced patient survival. Notably, ST6Gal-I upregulation in cancer cells conferred hallmark cancer stem-like cell (CSC) characteristics. Modulating ST6Gal-I expression in pancreatic and ovarian cancer cells directly altered CSC spheroid growth, and clonal variants with high ST6Gal-I activity preferentially survived in CSC culture. Primary ovarian cancer cells from patient ascites or solid tumors sorted for α2-6 sialylation grew as spheroids, while cells lacking α2-6 sialylation remained as single cells and lost viability. ST6Gal-I also promoted resistance to gemcitabine and enabled the formation of stably resistant colonies. Gemcitabine treatment of patient-derived xenograft tumors enriched for ST6Gal-I-expressing cells relative to pair-matched untreated tumors. ST6Gal-I also augmented tumor-initiating potential. In limiting dilution assays, subcutaneous tumor formation was inhibited by ST6Gal-I knockdown, whereas in a chemically induced tumor initiation model, mice with conditional ST6Gal-I overexpression exhibited enhanced tumorigenesis. Finally, we found that ST6Gal-I induced expression of the key tumor-promoting transcription factors, Sox9 and Slug. Collectively, this work highlighted a previously unrecognized role for a specific glycosyltransferase in driving a CSC state. Cancer Res; 76(13); 3978-88. ©2016 AACR.


Assuntos
Antígenos CD/metabolismo , Células-Tronco Neoplásicas/patologia , Neoplasias Ovarianas/patologia , Neoplasias Pancreáticas/patologia , Sialiltransferases/metabolismo , Fatores de Transcrição/metabolismo , Animais , Antígenos CD/genética , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Apoptose , Biomarcadores Tumorais , Estudos de Casos e Controles , Proliferação de Células , Estudos de Coortes , Feminino , Glicosilação , Humanos , Metástase Linfática , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Estadiamento de Neoplasias , Células-Tronco Neoplásicas/metabolismo , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Fenótipo , Prognóstico , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Sialiltransferases/genética , Fatores de Transcrição da Família Snail/genética , Fatores de Transcrição da Família Snail/metabolismo , Taxa de Sobrevida , Fatores de Transcrição/genética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Proteomics ; 16(3): 516-31, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26572242

RESUMO

We have applied a serologic proteomic workflow involving three complementary MS approaches to a tissue-specific Kras(G12D) -knockin mouse model of pancreatic cancer that consistently forms precancerous lesions by 4 months of age. The three proteomics applications were highly complementary and allowed us to survey the entire range of low to high molecular weight serologic proteins. Combined, we identified 121 (49↓, 72↑) unique and statistically relevant serologic biomarkers with 88% previously reported to be associated with cancer and 38% specifically correlated with pancreatic cancer. Four markers, lysozyme C2, cytokeratin 19, Serpina1A and Pcf11, were further verified by Western blotting. When applying systems analysis, the top-associated gene ontology functions were tied to wound healing, RXR signaling, growth, differentiation and innate immune activation through the JAK/STAT pathway. Upon further investigation of the apparent immune response using a multiplex cytokine screen, we found that IFN-γ, VEGF and GM-CSF were significantly increased in serum from the Kras(G12D) animals compared to littermate controls. By combining three complementary MS applications, we were able to survey the native intact peptidome and the global proteome in parallel, unveiling pathways that may be biologically relevant to promotion of pancreatic cancer progression and serologic markers of noninvasive early-stage neoplasia.


Assuntos
Biomarcadores Tumorais/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Pancreáticas/genética , Proteoma/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Animais , Biomarcadores Tumorais/sangue , Modelos Animais de Doenças , Progressão da Doença , Técnicas de Introdução de Genes , Fator Estimulador de Colônias de Granulócitos e Macrófagos/sangue , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Interferon gama/sangue , Interferon gama/genética , Queratina-19/sangue , Queratina-19/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Muramidase/sangue , Muramidase/genética , Neoplasias Pancreáticas/sangue , Neoplasias Pancreáticas/patologia , Proteoma/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/sangue , Transdução de Sinais , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Fator A de Crescimento do Endotélio Vascular/sangue , Fator A de Crescimento do Endotélio Vascular/genética , alfa 1-Antitripsina/sangue , alfa 1-Antitripsina/genética , Fatores de Poliadenilação e Clivagem de mRNA/sangue , Fatores de Poliadenilação e Clivagem de mRNA/genética
7.
Proc Natl Acad Sci U S A ; 113(1): E51-60, 2016 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-26699484

RESUMO

Epigenetic mechanisms play important regulatory roles in hematopoiesis and hematopoietic stem cell (HSC) function. Subunits of polycomb repressive complex 1 (PRC1), the major histone H2A ubiquitin ligase, are critical for both normal and pathological hematopoiesis; however, it is unclear which of the several counteracting H2A deubiquitinases functions along with PRC1 to control H2A ubiquitination (ubH2A) level and regulates hematopoiesis in vivo. Here we investigated the function of Usp16 in mouse hematopoiesis. Conditional deletion of Usp16 in bone marrow resulted in a significant increase of global ubH2A level and lethality. Usp16 deletion did not change HSC number but was associated with a dramatic reduction of mature and progenitor cell populations, revealing a role in governing HSC lineage commitment. ChIP- and RNA-sequencing studies in HSC and progenitor cells revealed that Usp16 bound to many important hematopoietic regulators and that Usp16 deletion altered the expression of genes in transcription/chromosome organization, immune response, hematopoietic/lymphoid organ development, and myeloid/leukocyte differentiation. The altered gene expression was partly rescued by knockdown of PRC1 subunits, suggesting that Usp16 and PRC1 counterbalance each other to regulate cellular ubH2A level and gene expression in the hematopoietic system. We further discovered that knocking down Cdkn1a (p21cip1), a Usp16 target and regulated gene, rescued the altered cell cycle profile and differentiation defect of Usp16-deleted HSCs. Collectively, these studies identified Usp16 as one of the histone H2A deubiquitinases, which coordinates with the H2A ubiquitin ligase PRC1 to regulate hematopoiesis, and revealed cell cycle regulation by Usp16 as key for HSC differentiation.


Assuntos
Hematopoese/fisiologia , Células-Tronco Hematopoéticas/citologia , Ubiquitina Tiolesterase/fisiologia , Proteases Específicas de Ubiquitina/fisiologia , Animais , Contagem de Células , Inibidor de Quinase Dependente de Ciclina p21/genética , Endopeptidases/genética , Endopeptidases/fisiologia , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Genes Letais , Hematopoese/genética , Histonas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Grupo Polycomb/genética , Proteínas do Grupo Polycomb/fisiologia , Transativadores , Ubiquitina Tiolesterase/genética , Proteases Específicas de Ubiquitina/genética
8.
Proteomics ; 12(22): 3393-402, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22997103

RESUMO

A number of reports have recently emerged with focus on extraction of proteins from formalin-fixed paraffin-embedded (FFPE) tissues for MS analysis; however, reproducibility and robustness as compared to flash frozen controls is generally overlooked. The goal of this study was to identify and validate a practical and highly robust approach for the proteomics analysis of FFPE tissues. FFPE and matched frozen pancreatic tissues obtained from mice (n = 8) were analyzed using 1D-nanoLC-MS(MS)(2) following work up with commercially available kits. The chosen approach for FFPE tissues was found to be highly comparable to that of frozen. In addition, the total number of unique peptides identified between the two groups was highly similar, with 958 identified for FFPE and 1070 identified for frozen, with protein identifications that corresponded by approximately 80%. This approach was then applied to archived human FFPE pancreatic cancer specimens (n = 11) as compared to uninvolved tissues (n = 8), where 47 potential pancreatic ductal adenocarcinoma markers were identified as significantly increased, of which 28 were previously reported. Further, these proteins share strongly overlapping pathway associations to pancreatic cancer that include estrogen receptor α. Together, these data support the validation of an approach for the proteomic analysis of FFPE tissues that is straightforward and highly robust, which can also be effectively applied toward translational studies of disease.


Assuntos
Carcinoma Ductal Pancreático/química , Pâncreas/química , Neoplasias Pancreáticas/química , Proteoma/análise , Proteômica/métodos , Sequência de Aminoácidos , Animais , Cromatografia Líquida , Análise por Conglomerados , Criopreservação/métodos , Formaldeído/química , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Inclusão em Parafina/métodos , Mapas de Interação de Proteínas , Proteoma/química , Proteômica/normas , Reprodutibilidade dos Testes , Estatísticas não Paramétricas , Biologia de Sistemas , Espectrometria de Massas em Tandem
9.
Blood ; 119(13): 2976-7, 2012 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-22461472

RESUMO

In this issue of Blood,Matsuura and colleagues provide evidence that loss of GMCSF signaling promotes leukemic progression in association with one of the most frequently observed cytogenetic abnormalities in AML, the t(8;21)(q22;q22) that generates the RUNX1-ETO fusion protein.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Leucemia Mieloide Aguda/genética , Transdução de Sinais/fisiologia , Translocação Genética , Animais , Humanos , Masculino
10.
Blood ; 119(6): 1522-31, 2012 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-21937700

RESUMO

The t(8;21) RUNX1-ETO translocation is one of the most frequent cytogenetic abnormalities in acute myeloid leukemia (AML). In RUNX1-ETO(+) patient samples, differing classes of activating c-KIT receptor tyrosine kinase mutations have been observed. The most common (12%-48%) involves mutations, such as D816V, which occur in the tyrosine kinase domain, whereas another involves mutations within exon 8 in a region mediating receptor dimerization (2%-13% of cases). To test whether distinct subtypes of activating c-KIT mutations differ in their leukemogenic potential in association with RUNX1-ETO, we used a retroviral transduction/transplantation model to coexpress RUNX1-ETO with either c-Kit(D814V) or c-Kit(T417IΔ418-419) in murine hematopoietic stem/progenitor cells used to reconstitute lethally irradiated mice. Analysis of reconstituted animals showed that RUNX1-ETO;c-Kit(D814V) coexpression resulted in 3 nonoverlapping phenotypes. In 45% of animals, a transplantable AML of relatively short latency and frequent granulocytic sarcoma was noted. Other mice exhibited a rapidly fatal myeloproliferative phenotype (35%) or a lethal, short-latency pre-B-cell leukemia (20%). In contrast, RUNX1-ETO;c-Kit(T417IΔ418-419) coexpression promoted exclusively AML in a fraction (51%) of reconstituted mice. These observations indicate that c-Kit(D814V) promotes a more varied and aggressive leukemic phenotype than c-Kit(T417IΔ418-419), which may be the result of differing potencies of the activating c-Kit alleles.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/genética , Leucemia Mieloide/genética , Mutação , Proteínas de Fusão Oncogênica/genética , Proteínas Proto-Oncogênicas c-kit/genética , Doença Aguda , Animais , Western Blotting , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Células Cultivadas , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Citometria de Fluxo , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Humanos , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patologia , Camundongos , Camundongos Endogâmicos C57BL , Células Mieloides/metabolismo , Células Mieloides/patologia , Células NIH 3T3 , Proteínas de Fusão Oncogênica/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Proteína 1 Parceira de Translocação de RUNX1 , Retroviridae/genética , Transdução Genética
11.
PLoS One ; 6(2): e16897, 2011 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-21379572

RESUMO

Harnessing the ability of cytotoxic T lymphocytes (CTLs) to recognize and eradicate tumor or pathogen-infected cells is a critical goal of modern immune-based therapies. Although multiple immunization strategies efficiently induce high levels of antigen-specific CTLs, the initial increase is typically followed by a rapid contraction phase resulting in a sharp decline in the frequency of functional CTLs. We describe a novel approach to immunotherapy based on a transplantation of low numbers of antigen-expressing hematopoietic stem cells (HSCs) following nonmyeloablative or partially myeloablative conditioning. Continuous antigen presentation by a limited number of differentiated transgenic hematopoietic cells results in an induction and prolonged maintenance of fully functional effector T cell responses in a mouse model. Recipient animals display high levels of antigen-specific CTLs four months following transplantation in contrast to dendritic cell-immunized animals in which the response typically declines at 4-6 weeks post-immunization. Majority of HSC-induced antigen-specific CD8+ T cells display central memory phenotype, efficiently kill target cells in vivo, and protect recipients against tumor growth in a preventive setting. Furthermore, we confirm previously published observation that high level engraftment of antigen-expressing HSCs following myeloablative conditioning results in tolerance and an absence of specific cytotoxic activity in vivo. In conclusion, the data presented here supports potential application of immunization by limited transplantation of antigen-expressing HSCs for the prevention and treatment of cancer and therapeutic immunization of chronic infectious diseases such as HIV-1/AIDS.


Assuntos
Antígenos/metabolismo , Transplante de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/metabolismo , Memória Imunológica/genética , Linfócitos T Citotóxicos/imunologia , Animais , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/transplante , Células Cultivadas , Galinhas/genética , Doenças Transmissíveis/imunologia , Doenças Transmissíveis/terapia , Terapia Genética/métodos , Células-Tronco Hematopoéticas/imunologia , Memória Imunológica/fisiologia , Imunoterapia Adotiva/métodos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neoplasias/imunologia , Neoplasias/terapia , Ovalbumina/genética , Ovalbumina/metabolismo , Linfócitos T Citotóxicos/metabolismo , Fatores de Tempo
12.
Proc Natl Acad Sci U S A ; 106(2): 552-7, 2009 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-19122139

RESUMO

Gene knockout experiments in mice have suggested a hierarchical model of early B cell commitment wherein E2A proteins (E47 and E12) activate early B cell factor (Ebf1), which in turn activates expression of the B cell commitment factor, Pax5. In IL-7 receptor alpha (IL-7Ralpha) knockout mice, B cell development is blocked before B-lineage commitment at the prepro-B cell stage in adult animals. In IL-7Ralpha(-/-) prepro-B cells, E47 is expressed and yet is insufficient to transcriptionally activate the putative downstream target gene, Ebf1. In this study, we show that further increases of E47 expression in IL-7Ralpha(-/-) prepro-B cells fails to activate Ebf1, but rather leads to a dramatic induction of the E2A inhibitory factors, Id2 and Id3. In contrast, enforced expression of Ebf1 in IL-7Ralpha(-/-) bone marrow potently down-regulates Id2 and Id3 mRNA expression and restores B cell differentiation in vivo. Down-regulation of both Id2 and Id3 during B cell specification is essential in that overexpression of either Id2 or Id3 in wild-type bone marrow blocks B cell specification at the prepro-B cell stage. Collectively, these studies suggest a model where Ebf1 induction specifies the B cell fate by dramatically increasing activity of E47 at the posttranslational level.


Assuntos
Linfócitos B/citologia , Regulação para Baixo , Proteína 2 Inibidora de Diferenciação/genética , Proteínas Inibidoras de Diferenciação/genética , Receptores de Interleucina-7/deficiência , Fatores de Transcrição TCF/fisiologia , Transativadores/fisiologia , Animais , Medula Óssea , Diferenciação Celular , Linhagem da Célula , Regulação da Expressão Gênica , Proteína 2 Inibidora de Diferenciação/fisiologia , Proteínas Inibidoras de Diferenciação/fisiologia , Interleucina-7/deficiência , Camundongos , Camundongos Knockout , Células Precursoras de Linfócitos B , RNA Mensageiro/análise , Fatores de Transcrição TCF/genética , Proteína 1 Semelhante ao Fator 7 de Transcrição
13.
J Gastrointest Surg ; 12(10): 1683-90, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18709425

RESUMO

BACKGROUND: The proteome varies with physiologic and disease states. Few studies have been reported that differentiate the proteome of those with pancreatic cancer. AIM: To apply proteomic-based technologies to body fluids. To differentiate pancreatic neoplasia from nonneoplastic pancreatic disease. METHODS: Samples from 50 patients (15 healthy (H), 24 cancer (Ca), 11 chronic pancreatitis (CP)) were prospectively collected and underwent analysis. A high-throughput method, using high-affinity solid lipophilic extraction resins, enriched low molecular weight proteins for extraction with a high-speed 200-Hz matrix-assisted laser desorption/ionization time-of-flight mass spectrometer (MALDI-MS; Bruker Ultraflex III). Samples underwent software processing with FlexAnalysis, Clinprot, MatLab, and Statistica (baseline, align, and normalize spectra). Nonparametric pairwise statistics, multidimensional scaling, hierarchical analysis, and leave-one-out cross validation completed the analysis. Sensitivity (sn) and specificity (sp) of group comparisons were determined. Two top-down-directed protein identification approaches were combined with MALDI-MS and tandem mass spectrometry to fully characterize the most significant protein biomarker. RESULTS: Using eight serum features, we differentiated Ca from H (sn 88%, sp 93%), Ca from CP (sn 88%, sp 30%), and Ca from both H and CP combined (sn 88%, sp 66%). In addition, nine features obtained from urine differentiated Ca from both H and CP combined with high efficiency (sn 90%, sp 90%). Interestingly, the plasma samples (considered by the Human Proteome Organization to be the preferred biological fluid) did not show significant differences. Multidimensional scaling indicated that markers from both serum and urine led to a highly effective clinical indicator of each specific disease state. CONCLUSIONS: The proteomic analysis of noninvasively acquired biological fluids provided a high level of predictability for diagnosing pancreatic cancer. While the proteomic analysis of serum was capable of screening individuals for pancreatic disease (i.e., CP and Ca vs. H), specific urine biomarkers further distinguished malignancy (Ca) from chronic inflammation (CP).


Assuntos
Biomarcadores Tumorais/análise , Neoplasias Pancreáticas/diagnóstico , Proteômica , Adulto , Idoso , Biomarcadores Tumorais/sangue , Biomarcadores Tumorais/urina , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Pancreatopatias/sangue , Pancreatopatias/diagnóstico , Pancreatopatias/urina , Neoplasias Pancreáticas/sangue , Neoplasias Pancreáticas/urina , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
14.
Leuk Res ; 32(7): 1101-11, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18037485

RESUMO

Inactivation of p15(Ink4b) expression by promoter hypermethylation occurs in up to 80% of acute myeloid leukemia (AML) cases and is particularly common in the FAB-M2 subtype of AML, which is characterized by the presence of the RUNX1-ETO translocation in 40% of cases. To establish whether the loss of p15(Ink4b) contributes to AML progression in association with RUNX1-ETO, we have expressed the RUNX1-ETO fusion protein from a retroviral vector in hematopoietic progenitor cells isolated from wild-type, p15(Ink4b) or p16(Ink4a) knockout bone marrow. Analysis of lethally irradiated recipient mice reconstituted with RUNX1-ETO-expressing cells showed that neither p15(Ink4b) or p16(Ink4a) loss significantly accelerated disease progression over the time period of one year post-transplantation. Loss of p15(Ink4b) alone resulted in increased myeloid progenitor cell frequencies in bone marrow by 10-month post-transplant and a 19-fold increase in the frequency of Lin(-)c-Kit(+)Sca-1(+) (LKS) cells that was not associated with expansion of long-term reconstituting HSC. These results strongly suggest that p15(Ink4b) loss must be accompanied by additional oncogenic changes for RUNX1-ETO-associated AML to develop.


Assuntos
Diferenciação Celular/fisiologia , Subunidade alfa 2 de Fator de Ligação ao Core/fisiologia , Inibidor de Quinase Dependente de Ciclina p15/fisiologia , Inibidor p16 de Quinase Dependente de Ciclina/fisiologia , Leucemia Mieloide Aguda/fisiopatologia , Animais , Sequência de Bases , Inibidor de Quinase Dependente de Ciclina p15/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , Primers do DNA , Leucemia Mieloide Aguda/patologia , Camundongos , Camundongos Endogâmicos C57BL
15.
Blood ; 111(3): 1567-74, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-17967943

RESUMO

The inversion of chromosome 16 in the inv(16)(p13q22) is one of the most frequent cytogenetic abnormalities observed in acute myeloid leukemia (AML). The inv(16) fuses the core binding factor (CBF) beta subunit with the coiled-coil rod domain of smooth muscle myosin heavy chain (SMMHC). Expression of CBFbeta-SMMHC in mice does not promote AML in the absence of secondary mutations. Patient samples with the inv(16) also possess mutually exclusive activating mutations in either N-RAS, K-RAS, or the receptor tyrosine kinases, c-KIT and FLT3, in almost 70% of cases. To test whether an activating mutation of FLT3 (FLT3-ITD) would cooperate with CBFbeta-SMMHC to promote AML, we coexpressed both mutations in hematopoietic progenitor cells used to reconstitute lethally irradiated mice. Analysis of transplanted animals showed strong selection for CBFbeta-SMMHC/FLT3-ITD-expressing cells in bone marrow and peripheral blood. Compared with animals transplanted with only CBFbeta-SMMHC-expressing cells, FLT3-ITD further restricted early myeloid differentiation and promoted peripheralization of primitive myeloblasts as early as 2.5 weeks after transplantation. FLT3-ITD also accelerated disease progression in all CBFbeta-SMMHC/FLT3-ITD-reconstituted animals, which died of a highly aggressive and transplantable AML within 3 to 5 months. These results indicate that FLT3-activating mutations can cooperate with CBFbeta-SMMHC in an animal model of inv(16)-associated AML.


Assuntos
Inversão Cromossômica/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Proteínas de Fusão Oncogênica/metabolismo , Tirosina Quinase 3 Semelhante a fms/metabolismo , Animais , Subunidade beta de Fator de Ligação ao Core/genética , Subunidade beta de Fator de Ligação ao Core/metabolismo , Progressão da Doença , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Leucemia Mieloide Aguda/genética , Linfopoese , Camundongos , Mutação/genética , Mielopoese , Proteínas de Fusão Oncogênica/genética , Miosinas de Músculo Liso/genética , Miosinas de Músculo Liso/metabolismo , Taxa de Sobrevida , Tirosina Quinase 3 Semelhante a fms/genética
16.
Cancer Res ; 67(17): 8121-30, 2007 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-17804724

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is one of the most fatal human malignancies, with an overall 5-year survival rate of <5%. Genetic analysis of PDAC patient samples has shown that specific disease-associated mutations are correlated with histologically defined stages of neoplastic progression in the ductal epithelium. Activating mutations in KRAS are almost uniformly present in early-stage disease, with subsequent inactivating mutations in p16(INK4A), p53, and SMAD4 occurring in more advanced lesions. In this study, we have tested whether the loss of Smad4 would cooperate with an activating Kras(G12D) mutation to promote progression to PDAC using the Pdx1-Cre transgenic system to activate Kras(G12D) and delete Smad4 in all pancreatic lineages including the ductal epithelium. Analysis of double-mutant mice showed that loss of Smad4 significantly accelerated the progression of pancreatic intraepithelial neoplasias (mPanIN) and promoted a high incidence of intraductal papillary mucinous neoplasia and active fibrosis compared with Pdx1-Cre;Kras(G12D) or Pdx1-Cre;Smad4(lox/lox) mice. Occasionally, double-mutant mice progressed to locally invasive PDAC with little evidence of metastases by 6 months of age and without the detectable loss of p53 or p16(Ink4A) expression or function. The loss of Smad4 only seemed to promote disease progression in the presence of the activated Kras(G12D) allele because we observed no abnormal pathology within the pancreata of 23 Pdx1-Cre;Smad4(lox/lox) animals that were analyzed up to 8 months of age. This indicates that Smad4 is dispensable for normal pancreatic development but is critical for at least partial suppression of multiple Kras(G12D)-dependent disease-associated phenotypes.


Assuntos
Carcinoma in Situ/genética , Genes ras , Neoplasias Pancreáticas/genética , Proteína Smad4/genética , Animais , Carcinoma in Situ/patologia , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Dilatação Patológica/genética , Dilatação Patológica/patologia , Progressão da Doença , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Transgênicos , Proteínas Mutantes/genética , Metástase Neoplásica , Neoplasias Pancreáticas/patologia , Pancreatite Crônica/genética , Pancreatite Crônica/patologia , Proteína Smad4/fisiologia
17.
Blood ; 109(5): 2190-7, 2007 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-17110463

RESUMO

Helios is a zinc-finger protein belonging to the Ikaros family of transcriptional regulators. It is expressed, along with Ikaros, throughout early stages of thymocyte development where it quantitatively associates with Ikaros through C-terminal zinc-finger domains that mediate heterodimerization between Ikaros family members. To understand the role of Helios in T-cell development, we used a retroviral vector to express full-length Helios or a Helios isoform that lacked the N-terminal DNA-binding domain in hematopoietic progenitor cells of reconstituted mice. Constitutive expression of full-length Helios resulted in an inhibition of T-cell development at the double-negative stage within the thymus. Although expression of the DNA-binding mutant of Helios did not contribute to developmental abnormalities at early times after transplantation, 60% of animals that expressed the Helios DNA-binding mutant developed an aggressive and transplantable T-cell lymphoma 4 to 10 months after transplantation. These results demonstrate a vital function for Helios in maintaining normal homeostasis of developing T cells and formally show that non-DNA-binding isoforms of Helios are lymphomagenic if aberrantly expressed within the T-cell lineage.


Assuntos
Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Proteínas de Ligação a DNA/metabolismo , Expressão Gênica , Linfoma de Células T/metabolismo , Linfoma de Células T/patologia , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Transformação Celular Neoplásica/genética , Células Cultivadas , Proteínas de Ligação a DNA/classificação , Proteínas de Ligação a DNA/genética , Células Matadoras Naturais/citologia , Células Matadoras Naturais/metabolismo , Linfoma de Células T/genética , Camundongos , Camundongos Endogâmicos C57BL , Mutação/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Timo/citologia , Timo/metabolismo , Fatores de Transcrição/classificação , Fatores de Transcrição/genética
18.
EMBO J ; 25(7): 1469-80, 2006 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-16525506

RESUMO

Homeobox (Hox) gene mutations and their altered expressions are frequently linked to human leukemia. Here, we report that transforming growth factor beta (TGFbeta)/bone morphogenetic protein (BMP) inhibits the bone marrow transformation capability of Hoxa9 and Nup98-Hoxa9, the chimeric fusion form of Hoxa9 identified in human acute myeloid leukemia (AML), through Smad4, the common Smad (Co-Smad) in the TGFbeta/BMP signaling pathway. Smad4 interacts directly with the homeodomain of Hoxa9 and blocks the ability of Nup98-Hoxa9 to bind DNA, thereby suppressing its ability to regulate downstream gene transcription. Mapping data revealed that the amino-terminus of Smad4 mediates this interaction and overexpression of the Hoxa9 interaction domain of Smad4 was sufficient to inhibit the enhanced serial replating ability of primary bone marrow cells induced by Nup98-Hoxa9. These studies establish a novel mechanism by which TGFbeta/BMP regulates hematopoiesis and suggest that modification of Hox DNA-binding activity may serve as a novel therapeutic intervention for those leukemias that involve deregulation of Hox.


Assuntos
Células da Medula Óssea/patologia , Proteínas Morfogenéticas Ósseas/metabolismo , Transformação Celular Neoplásica , Proteínas de Ligação a DNA/fisiologia , Proteínas de Homeodomínio/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Células da Medula Óssea/metabolismo , Células Cultivadas , Proteínas de Ligação a DNA/genética , Hematopoese , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Camundongos , Mutação , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Complexo de Proteínas Formadoras de Poros Nucleares/fisiologia , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/fisiologia , Ligação Proteica , Estrutura Terciária de Proteína , Transdução de Sinais , Proteína Smad4/metabolismo
19.
J Biol Chem ; 280(48): 40097-103, 2005 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-16199529

RESUMO

The inv(16) is one of the most frequent chromosomal translocations associated with acute myeloid leukemia (AML) and creates a chimeric fusion protein consisting of most of the runt-related X1 co-factor, core binding factor beta fused to the smooth muscle myosin heavy chain MYH11. Expression of the ARF tumor suppressor is regulated by runt-related X1, suggesting that the inv(16) fusion protein (IFP) may repress ARF expression. We established a murine bone marrow transplant model of the inv(16) in which wild type, Arf+/-, and Arf-/- bone marrow were engineered to express the IFP. IFP expression was sufficient to induce a myelomonocytic AML even when expressed in wild type bone marrow, yet removal of only a single allele of Arf greatly accelerated the disease, indicating that Arf is haploinsufficient for the induction of AML in the presence of the inv(16).


Assuntos
Leucemia Mieloide Aguda/genética , Proteínas de Fusão Oncogênica/fisiologia , Proteína Supressora de Tumor p14ARF/genética , Alelos , Animais , Células da Medula Óssea/citologia , Transplante de Medula Óssea , Inibidor p16 de Quinase Dependente de Ciclina , DNA/metabolismo , Citometria de Fluxo , Genes Reporter , Proteínas de Fluorescência Verde/metabolismo , Humanos , Leucemia Mieloide Aguda/metabolismo , Fígado/metabolismo , Camundongos , Camundongos Transgênicos , Mutagênese , Células NIH 3T3 , Proteínas de Fusão Oncogênica/metabolismo , Plasmídeos/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Recombinantes de Fusão/química , Baço/metabolismo , Fatores de Tempo , Transfecção , Translocação Genética , Proteína Supressora de Tumor p14ARF/metabolismo
20.
Blood ; 104(13): 3894-900, 2004 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-15328162

RESUMO

PU.1 is a member of the ETS family of transcription factors and is required for the development of multiple hematopoietic lineages. PU.1(-/-) mice die from hematopoietic failure at about embryonic day 18.5 (e18.5) and show a complete absence of B cells, mature T cells, and macrophages. This phenotype suggests that PU.1 may function at the level of the hematopoietic stem cell (HSC) or a multilineage progenitor. To investigate the role of PU.1 in the regulation of HSCs, PU.1(-/-) embryos were analyzed at various stages of embryonic development. The absolute number and frequency of HSCs were determined by flow cytometric analysis of c-Kit(+)Thy-1.1(lo)Lin(-)Sca-1(+) (KTLS) cells. We found that KTLS cells were absent or severely reduced in PU.1(-/-) fetal liver from e12.5 to e15.5. Progenitor cells with a c-Kit(+)Lin(-)AA4.1(+) and c-Kit(+)Lin(-)CD34(+) phenotype were also severely reduced. In addition, PU.1(-/-) fetal liver at e14.5 lacked common myeloid progenitors (CMPs) and granulocyte-macrophage progenitors (GMPs) but retained megakaryocyteerythroid progenitors (MEPs). Consistent with the loss of HSC activity, a 10-fold reduction in erythroid progenitors (mature erythroid burst-forming units [BFUEs]) was observed between e14.5 and e16.5. These data suggest that PU.1 plays an important role in the maintenance or expansion of HSC number in murine fetal liver.


Assuntos
Células-Tronco Hematopoéticas/fisiologia , Fígado/embriologia , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/fisiologia , Transativadores/deficiência , Transativadores/fisiologia , Animais , Cruzamentos Genéticos , Feto , Citometria de Fluxo , Fígado/citologia , Fígado/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Proto-Oncogênicas/genética , Transativadores/genética , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA