Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Blood ; 140(21): 2276-2289, 2022 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-36399071

RESUMO

Sickle cell disease (SCD) and ß-thalassemia are among the most common genetic disorders worldwide, affecting global health and mortality. Hemoglobin A2 (HbA2, α2δ2) is expressed at a low level in adult blood due to the lack of the Kruppel-like factor 1 (KLF1) binding motif in the δ-globin promoter region. However, HbA2 is fully functional as an oxygen transporter, and could be a valid antisickling agent in SCD, as well as a substitute for hemoglobin A in ß-thalassemia. We have previously demonstrated that KLF1-GATA1 fusion protein could interact with the δ-globin promoter and increase δ-globin expression in human primary CD34+ cells. We report the effects of 2 KLF1-GATA1 fusion proteins on hemoglobin expression, as well as SCD phenotypic correction in vitro and in vivo. Forced expression of KLF1-GATA1 fusion protein enhanced δ-globin gene and HbA2 expression, as well as reduced hypoxia-related sickling, in erythroid cells cultured from both human sickle CD34+ cells and SCD mouse hematopoietic stem cells (HSCs). The fusion proteins had no impact on erythroid cell differentiation, proliferation, and enucleation. Transplantation of highly purified SCD mouse HSCs expressing KLF1-GATA1 fusion protein into SCD mice lessened the severity of the anemia, reduced the sickling of red blood cells, improved SCD-related pathological alterations in spleen, kidney, and liver, and restored urine-concentrating ability in recipient mice. Taken together, these results indicate that the use of KLF1-GATA1 fusion constructs may represent a new gene therapy approach for hemoglobinopathies.


Assuntos
Anemia Falciforme , Proteínas Recombinantes de Fusão , Talassemia beta , Globinas delta , Animais , Humanos , Camundongos , Anemia Falciforme/genética , Anemia Falciforme/terapia , Antígenos CD34/metabolismo , Talassemia beta/genética , Globinas delta/genética , Fator de Transcrição GATA1/genética , Fenótipo , Proteínas Recombinantes de Fusão/uso terapêutico
2.
Int J Cancer ; 146(5): 1346-1358, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31241767

RESUMO

The olfactomedin 4 (OLFM4) gene has been analyzed as a tumor-suppressor gene and a putative biomarker in many cancers. In our study, we analyzed the relationship of OLFM4 expression with clinicopathological features and with CpG site methylation in the OLFM4 gene promoter region in human primary prostate adenocarcinoma. OLFM4 protein expression was significantly reduced in prostate cancer tissue compared to adjacent normal tissue and was further significantly reduced in more advanced cancers. Bioinformatic studies with clinical datasets revealed that primary prostate adenocarcinoma patients with reduced OLFM4 mRNA expression exhibited higher Gleason scores and higher preoperative serum prostate-specific antigen levels, as well as lower recurrence-free survival. Three of the eight CpG sites in the OLFM4 gene promoter region were hypermethylated in cancerous prostate cells compared to adjacent normal cells, and reduced methylation of eight CpG sites was associated with increased OLFM4 mRNA expression in RWPE1 and PC-3 cells. Furthermore, knockdown of OLFM4 gene expression was associated with enhanced epithelial-mesenchymal transition (EMT)-marker expression in RWPE immortalized normal prostate cells. In contrast, restoration of OLFM4 expression in PC-3 and DU145 prostate cancer cells lacking OLFM4 significantly inhibited both EMT-marker expression and tumor cell growth in in vitro and in vivo models, indicating that OLFM4 may play a tumor-suppressor role in inhibiting the EMT program, as well as tumor initiation and growth, in prostate cells. Taken together, these findings suggest that OLFM4 plays an important tumor-suppressor role in prostate cancer progression and might be useful as a novel candidate biomarker for prostate cancer.


Assuntos
Adenocarcinoma/genética , Regulação Neoplásica da Expressão Gênica , Fator Estimulador de Colônias de Granulócitos/genética , Neoplasias da Próstata/genética , Adenocarcinoma/mortalidade , Adenocarcinoma/patologia , Idoso , Animais , Linhagem Celular Tumoral , Proliferação de Células , Ilhas de CpG/genética , Metilação de DNA , Conjuntos de Dados como Assunto , Progressão da Doença , Intervalo Livre de Doença , Regulação para Baixo , Transição Epitelial-Mesenquimal/genética , Técnicas de Silenciamento de Genes , Fator Estimulador de Colônias de Granulócitos/metabolismo , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Regiões Promotoras Genéticas/genética , Próstata/patologia , Neoplasias da Próstata/mortalidade , Neoplasias da Próstata/patologia , Transdução de Sinais , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Blood Cells Mol Dis ; 77: 12-16, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30909074

RESUMO

BACKGROUND: Benign ethnic neutropenia (BEN), defined by neutrophil count <1.5 k/µL in the absence of other causes, is an asymptomatic condition more commonly observed in individuals of African ancestry. However, the natural history of this condition has been less well described. METHODS: Individuals with BEN were retrospectively identified by chart review or referral to hematology clinics. They were then invited to enroll in a prospective natural history study. Retrospective and prospective clinical and laboratory data were combined for descriptive analyses. FINDINGS: 46 participants, younger and older adults from 2 institutions, had BEN. Hypertension was reported in 30%, musculoskeletal disorders in 15%, and upper respiratory infection in 33% of these adults. Their leukopenia resulted from isolated neutropenia, ranging from 1000 and 1500 cells/µL. The severity of infections was mild and the frequency was similar to other healthy individuals in the ambulatory clinic. INTERPRETATION: In this group of BEN participants, their leukopenia was stable over time, and they had low rates of infections or common medical disorders, confirming the benign nature of this condition. The presence of BEN in children, younger adults, and older adults suggest a hereditary pattern for BEN.


Assuntos
População Negra , Neutropenia/epidemiologia , Adolescente , Adulto , Idoso , Criança , Comorbidade , Feminino , Humanos , Contagem de Leucócitos , Masculino , Pessoa de Meia-Idade , Neutropenia/complicações , Neutropenia/diagnóstico , Neutropenia/terapia , Neutrófilos , Vigilância da População , Estudos Retrospectivos , Adulto Jovem
4.
Am J Physiol Cell Physiol ; 315(4): C494-C501, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-29949402

RESUMO

Neutrophils increase production of reactive oxygen species, including superoxide, hydrogen peroxide (H2O2), and hydroxyl radical, to destroy invading microorganisms under pathological conditions. Conversely, oxidative stress conditions, such as the presence of H2O2, induce neutrophil apoptosis, which helps to remove neutrophils after inflammation. However, the detailed molecular mechanisms that are involved in the latter process have not been elucidated. In this study, we investigated the potential role of olfactomedin 4 (Olfm4) in H2O2-induced superoxide production and apoptosis in mouse neutrophils. We have demonstrated that Olfm4 is not required for maximal-dosage PMA- and Escherichia coli bacteria-induced superoxide production, but Olfm4 contributes to suboptimal-dosage PMA- and H2O2-induced superoxide production. Using an NADPH oxidase inhibitor and gp91phox-deficient mouse neutrophils, we found that NAPDH oxidase was required for PMA-stimulated superoxide production and that Olfm4 mediated H2O2-induced superoxide production through NADPH oxidase, in mouse neutrophils. We have shown that neutrophils from Olfm4-deficient mice exhibited reduced H2O2-induced apoptosis compared with neutrophils from wild-type mice. We also demonstrated that neutrophils from Olfm4-deficient mice exhibited reduced H2O2-stimulated mitochondrial damage and membrane permeability, and as well as reduced caspase-3 and caspase-9 activity, compared with neutrophils from wild-type mice. Moreover, the cytoplasmic translocation of the proapoptotic mitochondrial proteins Omi/HtrA2 and Smac/DIABLO in response to H2O2 was reduced in neutrophils from Olfm4-deficient mice compared with neutrophils from wild-type mice. Our study demonstrates that Olfm4 contributes to H2O2-induced NADPH oxidase activation and apoptosis in mouse neutrophils. Olfactomedin 4 might prove to be a potential target for future studies on inflammatory neutrophil biology and for inflammatory disease treatment.


Assuntos
Apoptose/fisiologia , Glicoproteínas/metabolismo , Peróxido de Hidrogênio/farmacologia , NADPH Oxidases/metabolismo , Neutrófilos/metabolismo , Animais , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Caspase 9/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Mitocondriais/metabolismo , Neutrófilos/efeitos dos fármacos , Oxirredução/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Superóxidos/metabolismo
7.
J Biol Chem ; 291(16): 8549-64, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26895964

RESUMO

Monocyte migration requires the dynamic redistribution of integrins through a regulated endo-exocytosis cycle, but the complex molecular mechanisms underlying this process have not been fully elucidated. Glia maturation factor-γ (GMFG), a novel regulator of the Arp2/3 complex, has been shown to regulate directional migration of neutrophils and T-lymphocytes. In this study, we explored the important role of GMFG in monocyte chemotaxis, adhesion, and ß1-integrin turnover. We found that knockdown of GMFG in monocytes resulted in impaired chemotactic migration toward formyl-Met-Leu-Phe (fMLP) and stromal cell-derived factor 1α (SDF-1α) as well as decreased α5ß1-integrin-mediated chemoattractant-stimulated adhesion. These GMFG knockdown impaired effects could be reversed by cotransfection of GFP-tagged full-length GMFG. GMFG knockdown cells reduced the cell surface and total protein levels of α5ß1-integrin and increased its degradation. Importantly, we demonstrate that GMFG mediates the ubiquitination of ß1-integrin through knockdown or overexpression of GMFG. Moreover, GMFG knockdown retarded the efficient recycling of ß1-integrin back to the plasma membrane following normal endocytosis of α5ß1-integrin, suggesting that the involvement of GMFG in maintaining α5ß1-integrin stability may occur in part by preventing ubiquitin-mediated degradation and promoting ß1-integrin recycling. Furthermore, we observed that GMFG interacted with syntaxin 4 (STX4) and syntaxin-binding protein 4 (STXBP4); however, only knockdown of STXBP4, but not STX4, reduced monocyte migration and decreased ß1-integrin cell surface expression. Knockdown of STXBP4 also substantially inhibited ß1-integrin recycling in human monocytes. These results indicate that the effects of GMFG on monocyte migration and adhesion probably occur through preventing ubiquitin-mediated proteasome degradation of α5ß1-integrin and facilitating effective ß1-integrin recycling back to the plasma membrane.


Assuntos
Movimento Celular/fisiologia , Fator de Maturação da Glia/metabolismo , Integrina beta1/metabolismo , Monócitos/metabolismo , Movimento Celular/efeitos dos fármacos , Quimiocina CXCL12/farmacologia , Técnicas de Silenciamento de Genes , Fator de Maturação da Glia/genética , Humanos , Integrina alfa5beta1/genética , Integrina alfa5beta1/metabolismo , Integrina beta1/genética , Monócitos/citologia , N-Formilmetionina Leucil-Fenilalanina/farmacologia , Proteínas Qa-SNARE/genética , Proteínas Qa-SNARE/metabolismo , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
8.
Cancer Metastasis Rev ; 35(2): 201-12, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27178440

RESUMO

Olfactomedin 4 (OLFM4) is an olfactomedin domain-containing glycoprotein. Multiple signaling pathways and factors, including NF-κB, Wnt, Notch, PU.1, retinoic acids, estrogen receptor, and miR-486, regulate its expression. OLFM4 interacts with several other proteins, such as gene associated with retinoic-interferon-induced mortality 19 (GRIM-19), cadherins, lectins, nucleotide oligomerization domain-1 (NOD1) and nucleotide oligomerization domain-2 (NOD2), and cathepsins C and D, known to regulate important cellular functions. Recent investigations using Olfm4-deficient mouse models have provided important clues about its in vivo biological functions. Olfm4 inhibited Helicobacter pylori-induced NF-κB pathway activity and inflammation and facilitated H. pylori colonization in the mouse stomach. Olfm4-deficient mice exhibited enhanced immunity against Escherichia coli and Staphylococcus aureus infection. Olfm4 deletion in a chronic granulomatous disease mouse model rescued them from S. aureus infection. Olfm4 deletion in mice treated with azoxymethane/dextran sodium sulfate led to robust intestinal inflammation and intestinal crypt hyperplasia. Olfm4 deletion in Apc (Min/+) mice promoted intestinal polyp formation as well as adenocarcinoma development in the distal colon. Further, Olfm4-deficient mice spontaneously developed prostatic epithelial lesions as they age. OLFM4 expression is correlated with cancer differentiation, stage, metastasis, and prognosis in a variety of cancers, suggesting its potential clinical value as an early-stage cancer marker or a therapeutic target. Collectively, these data suggest that OLFM4 plays important roles in innate immunity against bacterial infection, gastrointestinal inflammation, and cancer. In this review, we have summarized OLFM4's initial characterization, expression, regulation, protein interactions, and biological functions.


Assuntos
Regulação da Expressão Gênica , Fator Estimulador de Colônias de Granulócitos/genética , Imunidade Inata/genética , Inflamação/genética , Inflamação/imunologia , Neoplasias/genética , Neoplasias/imunologia , Animais , Infecções Bacterianas/genética , Infecções Bacterianas/imunologia , Infecções Bacterianas/metabolismo , Proteínas de Transporte , Fator Estimulador de Colônias de Granulócitos/química , Fator Estimulador de Colônias de Granulócitos/metabolismo , Humanos , Inflamação/metabolismo , MicroRNAs/genética , NF-kappa B/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Ligação Proteica , Transdução de Sinais
9.
J Am Soc Nephrol ; 27(9): 2576-95, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27178804

RESUMO

Significant disparities in CKD rates and outcomes exist between black and white Americans. Health disparities are defined as health differences that adversely affect disadvantaged populations, on the basis of one or more health outcomes. CKD is the complex result of genetic and environmental factors, reflecting the balance of nature and nurture. Social determinants of health have an important role as environmental components, especially for black populations, who are disproportionately disadvantaged. Understanding the social determinants of health and appreciating the underlying differences associated with meaningful clinical outcomes may help nephrologists treat all their patients with CKD in an optimal manner. Altering the social determinants of health, although difficult, may embody important policy and research efforts, with the ultimate goal of improving outcomes for patients with kidney diseases, and minimizing the disparities between groups.


Assuntos
Disparidades nos Níveis de Saúde , Grupos Raciais , Insuficiência Renal Crônica/epidemiologia , Determinantes Sociais da Saúde , Acessibilidade aos Serviços de Saúde , Humanos , Modelos Teóricos , Insuficiência Renal Crônica/etnologia , Fatores Socioeconômicos
11.
J Urol ; 196(5): 1342-1349, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27341750

RESUMO

PURPOSE: Lower urinary tract symptoms are common in the United States population, leading to significant economic, quality of life and public health issues. The burden will increase as the population ages, and risk factors for lower urinary tract symptoms, including diabetes and obesity, remain highly prevalent. Improving clinical management and establishing the knowledge base to prevent lower urinary tract symptoms will require a comprehensive research approach that examines factors beyond the lower urinary tract. While the study of extra-lower urinary tract factors has increased recently, current urological research does not systematically account for the broad set of potential contributing factors spanning biological, behavioral, psychological/executive function and sociocultural factors. A comprehensive assessment of potential contributors to risk, treatment response and progression is necessary to reduce the burden of this condition in the United States. MATERIALS AND METHODS: We considered challenges to continuing the predominantly lower urinary tract dysfunction centric approach that has dominated previous research of lower urinary tract symptoms. RESULTS: We developed a new, comprehensive framework for urology research that includes a broader set of potential factors contributing to lower urinary tract symptoms. This framework aims to broaden research to consider a comprehensive set of potential contributing factors and to engage a broad range of researchers in the investigation of as many extra-lower urinary tract factors as possible, with the goal of improving clinical care and prevention. CONCLUSIONS: We propose a new framework for future urology research, which should help to reduce the medical and economic burden of lower urinary tract symptoms in the United States population.


Assuntos
Sintomas do Trato Urinário Inferior , Pesquisa Biomédica , Humanos , Urologia
12.
Blood ; 124(7): 1146-56, 2014 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-24914133

RESUMO

Hydroxyurea (HU) is effectively used in the management of ß-hemoglobinopathies by augmenting the production of fetal hemoglobin (HbF). However, the molecular mechanisms underlying HU-mediated HbF regulation remain unclear. We previously reported that overexpression of the HU-induced SAR1 gene closely mimics the known effects of HU on K562 and CD34(+) cells, including γ-globin induction and cell-cycle regulation. Here, we show that HU stimulated nuclear factor-κB interaction with its cognate-binding site on the SAR1 promoter to regulate transcriptional expression of SAR1 in K562 and CD34(+) cells. Silencing SAR1 expression not only significantly lowered both basal and HU-elicited HbF production in K562 and CD34(+) cells, but also significantly reduced HU-mediated S-phase cell-cycle arrest and apoptosis in K562 cells. Inhibition of c-Jun N-terminal kinase (JNK)/Jun phosphorylation and silencing of Giα expression in SAR1-transfected K562 and CD34(+) cells reduced both γ-globin expression and HbF level, indicating that activation of Giα/JNK/Jun proteins is required for SAR1-mediated HbF induction. Furthermore, reciprocal coimmunoprecipitation assays revealed an association between forcibly expressed SAR1 and Giα2 or Giα3 proteins in both K562 and nonerythroid cells. These results indicate that HU induces SAR1, which in turn activates γ-globin expression, predominantly through the Giα/JNK/Jun pathway. Our findings identify SAR1 as an alternative therapeutic target for ß-globin disorders.


Assuntos
Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Hidroxiureia/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , gama-Globinas/metabolismo , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Sítios de Ligação/genética , Western Blotting , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Células Cultivadas , Hemoglobina Fetal/genética , Hemoglobina Fetal/metabolismo , Expressão Gênica/efeitos dos fármacos , Humanos , Células K562 , Proteínas Monoméricas de Ligação ao GTP/genética , NF-kappa B/metabolismo , Regiões Promotoras Genéticas/genética , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Proteínas Elk-1 do Domínio ets/metabolismo , gama-Globinas/genética
13.
Br J Haematol ; 170(6): 781-7, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26104837

RESUMO

HbA2 , a tetramer of α- and δ-globin chains, provides a diagnostic clue to the presence of ß-thalassaemia trait. This minor haemoglobin, which forms about 2-3% of the total, has no known physiological role, but has the interesting property of preventing polymerization of deoxy-sickle haemoglobin. If it were possible to increase the level of HbA2 sufficiently it could have a benefit in sickle cell disease similar to that of foetal haemoglobin. Moreover, HbA2 is present in all erythrocytes, an advantage not found with foetal haemoglobin, which is heterocellularly expressed. The molecular basis of HbA2 gene (HBD) expression is partially understood, and with new molecular tools, it might be possible to induce levels of HbA2 that could be clinically important. However, high concentrations of this positively charged haemoglobin might damage the erythrocyte membrane; also, the reciprocal relationship of δ- and γ-globin gene (HBD and HBG1/2, respectively) expression might negate any benefit of increasing transcription of the former.


Assuntos
Anemia Falciforme/etiologia , Hemoglobina A2/fisiologia , Anemia Falciforme/tratamento farmacológico , Animais , Evolução Molecular , Hemoglobina A2/química , Humanos , Biossíntese de Proteínas
14.
BMC Med Genet ; 16: 103, 2015 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-26686224

RESUMO

BACKGROUND: Hyperuricemia and associated cardio-metabolic disorders are more prevalent in African Americans than in European Americans. We used genome-wide admixture mapping and association testing to identify loci with ancestry effects on serum uric acid levels. METHODS: We analyzed 1,976 African Americans from Washington, D.C, including 1,322 individuals from 328 pedigrees and 654 unrelated individuals, enrolled in the Howard University Family Study. We performed admixture mapping and genome-wide association testing using ~800 k autosomal single-nucleotide polymorphisms (SNPs). We performed fine mapping by dense genotyping. We assessed functionality by a combination of bioinformatic annotation, reporter gene assays, and gel shift experiments. We also analyzed 12,641 individuals enrolled in the National Health and Nutrition Examination Survey. RESULTS: We detected a genome-wide significant locus on chromosome 11p15.4 at which serum uric acid levels increased with increasing African ancestry, independent of kidney function. Fine-mapping identified two independent signals in the ß-globin locus. The ancestral allele at SNP rs2855126, located upstream of the hemoglobin, gamma A gene HBG1, was associated with increased serum uric acid levels and higher expression of a reporter gene relative to the derived allele. Hyperuricemia was associated with increased risk of hypertension in 3,767 African Americans (Odds Ratio = 2.48, p = 2.71 × 10(-19)). CONCLUSIONS: Given that increased expression of γ-globin leads to increased levels of fetal hemoglobin which confers protection against malaria, we hypothesize that evolution in Africa of protection against malaria may have occurred at the cost of increased serum uric acid levels, contributing to the high rates of hyperuricemia and associated cardio-metabolic disorders observed in African Americans.


Assuntos
Negro ou Afro-Americano/genética , Hipertensão/etnologia , Ácido Úrico/sangue , gama-Globinas/genética , Adulto , Evolução Biológica , População Negra/genética , Mapeamento Cromossômico , Feminino , Frequência do Gene , Estudo de Associação Genômica Ampla/métodos , Genótipo , Geografia , Células HEK293 , Humanos , Células K562 , Masculino , Pessoa de Meia-Idade , Inquéritos Nutricionais/estatística & dados numéricos , Polimorfismo de Nucleotídeo Único , Estados Unidos/epidemiologia , População Branca/genética
15.
Blood ; 121(16): 3216-27, 2013 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-23327923

RESUMO

Human erythropoiesis is a dynamic and complex multistep process involving differentiation of early erythroid progenitors into enucleated RBCs. The mechanisms underlying erythropoiesis still remain incompletely understood. We previously demonstrated that erythropoietin-stimulated clone-1, which is selectively expressed in normal human erythroid-lineage cells, shares 99.5% identity with malignant fibrous histiocytoma-amplified sequences with leucine-rich tandem repeats 1 (MASL1). In this study, we hypothesized that the MASL1 gene plays a role in erythroid differentiation, and used a human erythroid cell culture system to explore this concept. MASL1 mRNA and protein expression levels were significantly increased during the erythroid differentiation of CD34(+) cells following erythropoietin (EPO) treatment. Conversely, MASL1 knockdown reduced erythroid differentiation in EPO-treated CD34(+) cells. In addition, MASL1 knockdown interrupted the Raf/MEK/ERK signaling pathway in CD34(+) cells. MASL1 mutant-transfected CD34(+) cells also showed decreased erythroid differentiation. Furthermore, inhibition of the SH3 domain of Son of Sevenless, which is an upstream adapter protein in EPO-induced erythroid differentiation, also reduced MASL1 expression and phosphorylation of Raf/MEK/ERK kinases that consequently reduced erythroid differentiation of EPO-induced CD34(+) cells. Importantly, we also demonstrated that MASL1 interacts physically with Raf1. Taken together, our data provide novel insights into MASL1 regulation of erythropoiesis through the Raf/MEK/ERK pathway.


Assuntos
Antígenos CD34/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Células Eritroides/citologia , Eritropoese , Eritropoetina/metabolismo , Sistema de Sinalização das MAP Quinases , Proteínas Oncogênicas/metabolismo , Quinases raf/metabolismo , Pontos de Checagem do Ciclo Celular , Proteínas de Ciclo Celular/genética , Proliferação de Células , Células Cultivadas , Proteínas de Ligação a DNA/genética , Eritrócitos/citologia , Eritrócitos/metabolismo , Células Eritroides/metabolismo , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/metabolismo , Técnicas de Silenciamento de Genes , Granulócitos/citologia , Granulócitos/metabolismo , Humanos , Proteínas Oncogênicas/genética , RNA Mensageiro/genética , Proteínas Son Of Sevenless/metabolismo , Regulação para Cima
16.
J Immunol ; 190(12): 6093-103, 2013 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-23677465

RESUMO

TLR4 signaling must be tightly regulated to provide both effective immune protection and avoid inflammation-induced pathology. Thus, the mechanisms that negatively regulate the TLR4-triggered inflammatory response are of particular importance. Glia maturation factor-γ (GMFG), a novel actin depolymerization factor/cofilin superfamily protein that is expressed in inflammatory cells, has been implicated in mediating neutrophil and T cell migration, but its function in macrophage immune response remains unclear. In the current study, the role of GMFG in the LPS-induced TLR4-signaling pathway was investigated in THP-1 macrophages and human primary macrophages. LPS stimulation of macrophages decreased GMFG mRNA and protein expression. We show that GMFG negatively regulates LPS-induced activation of NF-κB-, MAPK-, and IRF3-signaling pathways and subsequent production of proinflammatory cytokines and type I IFN in human macrophages. We found that endogenous GMFG localized within early and late endosomes. GMFG knockdown delayed LPS-induced TLR4 internalization and caused prolonged TLR4 retention at the early endosome, suggesting that TLR4 transport from early to late endosomes is interrupted, which may contribute to enhanced LPS-induced TLR4 signaling. Taken together, our findings suggest that GMFG functions as a negative regulator of TLR4 signaling by facilitating TLR4 endocytic trafficking in macrophages.


Assuntos
Fator de Maturação da Glia/metabolismo , Macrófagos/metabolismo , Transdução de Sinais/fisiologia , Receptor 4 Toll-Like/metabolismo , Citometria de Fluxo , Imunofluorescência , Fator de Maturação da Glia/imunologia , Humanos , Immunoblotting , Macrófagos/imunologia , Microscopia Confocal , Reação em Cadeia da Polimerase , Transporte Proteico/fisiologia , Interferência de RNA , Receptor 4 Toll-Like/imunologia
20.
Am J Pathol ; 183(4): 1329-38, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24070418

RESUMO

The olfactomedin 4 (OLFM4) gene is located on chromosome 13q14.3, which frequently is deleted in human prostate cancer. However, direct genetic evidence of OLFM4 gene alteration in human prostate cancer has not yet been obtained. In this study, we investigated the genetics, protein expression, and functions of the OLFM4 gene in human prostate cancer. We found overall 25% deletions within the OLFM4 gene in cancerous epithelial cells compared with adjacent normal epithelial cells that were microdissected from 31 prostate cancer specimens using laser-capture microdissection and genomic DNA sequencing. We found 28% to 45% hemizygous and 15% to 57% homozygous deletions of the OLFM4 gene via fluorescence in situ hybridization analysis from 44 different prostate cancer patient samples. Moreover, homozygous deletion of the OLFM4 gene significantly correlated with advanced prostate cancer. By using immunohistochemical analysis of 162 prostate cancer tissue array samples representing a range of Gleason scores, we found that OLFM4 protein expression correlated inversely with advanced prostate cancer, consistent with the genetic results. We also showed that a truncated mutant of OLFM4 that lacks the olfactomedin domain eliminated suppression of PC-3 prostate cancer cell growth. Together, our findings indicate that OLFM4 is a novel candidate tumor-suppressor gene for chromosome 13q and may shed new light on strategies that could be used for the diagnosis, prognosis, and treatment of prostate cancer patients.


Assuntos
Progressão da Doença , Deleção de Genes , Fator Estimulador de Colônias de Granulócitos/genética , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Autofagia/genética , Sequência de Bases , Catepsina D/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Genoma Humano/genética , Fator Estimulador de Colônias de Granulócitos/química , Fator Estimulador de Colônias de Granulócitos/metabolismo , Homozigoto , Humanos , Hibridização in Situ Fluorescente , Microdissecção e Captura a Laser , Masculino , Dados de Sequência Molecular , Proteínas Mutantes/metabolismo , Estadiamento de Neoplasias , Polimorfismo de Nucleotídeo Único/genética , Neoplasias da Próstata/enzimologia , Estrutura Terciária de Proteína , Análise Serial de Tecidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA