Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 13(1): 10290, 2023 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-37357228

RESUMEN

Olfactomedin4 (Olfm4) is expressed in normal mouse prostate. However, Olfm4+ cells in the murine prostate have not been well characterized. In this study, we generated an Olfm4eGFP reporter mouse line with C57BL/6 mice and investigated the distribution of Olfm4/eGFP-expressing cells during postnatal development from P1, P7, P14, P20, P42, P56 to adult male mouse prostate and urethral tube. We observed Olfm4/eGFP expression in urogenital and prostatic epithelial cells during early postnatal development, which persisted into adulthood in urethral-tube and anterior-prostate (AP) epithelium. We found Olfm4+ cells are E-cadherin+/CD44+/Foxa1+ and some of subpopulation are Ck8+/Ck5+/Sca-1-/Ck4-/Syn- in the adult mouse AP epithelium. Functional studies of single-cell preparations of Olfm4/eGFP-expressing cells isolated from adult Olfm4eGFP mouse prostate demonstrated that Olfm4+ cells can grow and form colonies, spheres, or organoids in culture. Bioinformatic analysis of Olfm4+ cells using single-cell RNA sequencing meta data in adult mouse urethra (GSE145865) identified upregulation of genes related to cell and tissue migration and development, as well as upregulation of xenobiotic metabolism signaling pathways. In conclusion, Olfm4eGFP mouse is a novel model to further study Olfm4's biological functions and Olfm4+ cells may contribute importantly to cellular processes supporting development and homeostasis of the epithelium in murine prostate and urethral tube.


Asunto(s)
Glicoproteínas , Próstata , Ratones , Masculino , Animales , Próstata/metabolismo , Ratones Endogámicos C57BL , Epitelio/metabolismo , Glicoproteínas/metabolismo , Factor Estimulante de Colonias de Granulocitos/metabolismo
3.
Blood ; 140(21): 2276-2289, 2022 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-36399071

RESUMEN

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.


Asunto(s)
Anemia de Células Falciformes , Proteínas Recombinantes de Fusión , Talasemia beta , Globinas delta , Animales , Humanos , Ratones , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/terapia , Antígenos CD34/metabolismo , Talasemia beta/genética , Globinas delta/genética , Factor de Transcripción GATA1/genética , Fenotipo , Proteínas Recombinantes de Fusión/uso terapéutico
4.
Sci Rep ; 10(1): 21924, 2020 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-33318499

RESUMEN

Olfactomedin 4 (OLFM4) is expressed in normal prostate epithelial cells and immortalized normal human prostate epithelial cells (RWPE1), but the identity of OLFM4-expressing cells within these populations and OLFM4's physiological functions in these cells have not been elucidated. Using single-cell RNA sequencing analysis, we found here that OLFM4 was expressed in multiple stem/progenitor-like cell populations in both the normal prostate epithelium and RWPE1 cells and was frequently co-expressed with KRT13 and LY6D in RWPE1 cells. Functionally, OLFM4-knockout RWPE1 cells exhibited enhanced proliferation of the stem/progenitor-like cell population, shifts stem/progenitor-like cell division to favor symmetric division and differentiated into higher levels PSA expression cells in organoid assays compared with OLFM4-wild RWPE1 cells. Bulk-cell RNA sequencing analysis pinpointed that cMYC expression were enhanced in the OLFM4-knockout RWPE1 cells compared with OLFM4-wild cells. Molecular and signaling pathway studies revealed an increase in the WNT/APC/MYC signaling pathway gene signature, as well as that of MYC target genes that regulate multiple biological processes, in OLFM4-knockout RWPE1 cells. These findings indicated that OLFM4 is co-expressed with multiple stem/progenitor cell marker genes in prostate epithelial cells and acts as a novel mediator in prostate stem/progenitor cell proliferation and differentiation.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Regulación de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos/biosíntesis , Próstata/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Células Madre/metabolismo , Línea Celular Transformada , Factor Estimulante de Colonias de Granulocitos/genética , Humanos , Masculino , Próstata/citología , Proteínas Proto-Oncogénicas c-myc/genética , Células Madre/citología
5.
Int J Cancer ; 146(5): 1346-1358, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31241767

RESUMEN

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.


Asunto(s)
Adenocarcinoma/genética , Regulación Neoplásica de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos/genética , Neoplasias de la Próstata/genética , Adenocarcinoma/mortalidad , Adenocarcinoma/patología , Anciano , Animales , Línea Celular Tumoral , Proliferación Celular , Islas de CpG/genética , Metilación de ADN , Conjuntos de Datos como Asunto , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Regulación hacia Abajo , Transición Epitelial-Mesenquimal/genética , Técnicas de Silenciamiento del Gen , Factor Estimulante de Colonias de Granulocitos/metabolismo , Humanos , Masculino , Ratones , Persona de Mediana Edad , Regiones Promotoras Genéticas/genética , Próstata/patología , Neoplasias de la Próstata/mortalidad , Neoplasias de la Próstata/patología , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
7.
J Biol Chem ; 291(16): 8549-64, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26895964

RESUMEN

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.


Asunto(s)
Movimiento Celular/fisiología , Factor de Maduración de la Glia/metabolismo , Integrina beta1/metabolismo , Monocitos/metabolismo , Movimiento Celular/efectos de los fármacos , Quimiocina CXCL12/farmacología , Técnicas de Silenciamiento del Gen , Factor de Maduración de la Glia/genética , Humanos , Integrina alfa5beta1/genética , Integrina alfa5beta1/metabolismo , Integrina beta1/genética , Monocitos/citología , N-Formilmetionina Leucil-Fenilalanina/farmacología , Proteínas Qa-SNARE/genética , Proteínas Qa-SNARE/metabolismo , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
8.
Sci Rep ; 5: 16974, 2015 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-26581960

RESUMEN

Loss of olfactomedin 4 (OLFM4) gene expression is associated with the progression of human prostate cancer, but its role and the molecular mechanisms involved in this process have not been completely understood. In this study, we found that Olfm4-knockout mice developed prostatic intraepithelial neoplasia and prostatic adenocarcinoma. Importantly, we found that the hedgehog-signaling pathway was significantly upregulated in the Olfm4-knockout mouse model. We also found that restoration of OLFM4 in human prostate-cancer cells that lack OLFM4 expression significantly downregulated hedgehog signaling-pathway component expression. Furthermore, we demonstrated that the OLFM4 protein interacts with sonic hedgehog protein, as well as significantly inhibits GLI-reporter activity. Bioinformatic and immunohistochemistry analyses revealed that decreased OLFM4 and increased SHH expression was significantly associated with advanced human prostate cancer. Thus, olfactomedin 4 appears to play a critical role in regulating progression of prostate cancer, and has potential as a new biomarker for prostate cancer.


Asunto(s)
Progresión de la Enfermedad , Glicoproteínas/deficiencia , Factor Estimulante de Colonias de Granulocitos/deficiencia , Proteínas Hedgehog/metabolismo , Neoplasias de la Próstata/patología , Transducción de Señal , Regulación hacia Arriba , Animales , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Regulación hacia Abajo , Células Epiteliales/patología , Epitelio/metabolismo , Epitelio/patología , Regulación Neoplásica de la Expresión Génica , Genes Reporteros , Glicoproteínas/metabolismo , Factor Estimulante de Colonias de Granulocitos/metabolismo , Humanos , Masculino , Ratones Noqueados , Neoplasias de la Próstata/genética , Unión Proteica
9.
Blood ; 124(7): 1146-56, 2014 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-24914133

RESUMEN

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.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Hidroxiurea/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , gamma-Globinas/metabolismo , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Sitios de Unión/genética , Western Blotting , Puntos de Control del Ciclo Celular/efectos de los fármacos , Células Cultivadas , Hemoglobina Fetal/genética , Hemoglobina Fetal/metabolismo , Expresión Génica/efectos de los fármacos , Humanos , Células K562 , Proteínas de Unión al GTP Monoméricas/genética , FN-kappa B/metabolismo , Regiones Promotoras Genéticas/genética , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Proteína Elk-1 con Dominio ets/metabolismo , gamma-Globinas/genética
10.
Am J Pathol ; 183(4): 1329-38, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24070418

RESUMEN

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.


Asunto(s)
Progresión de la Enfermedad , Eliminación de Gen , Factor Estimulante de Colonias de Granulocitos/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Autofagia/genética , Secuencia de Bases , Catepsina D/metabolismo , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Genoma Humano/genética , Factor Estimulante de Colonias de Granulocitos/química , Factor Estimulante de Colonias de Granulocitos/metabolismo , Homocigoto , Humanos , Hibridación Fluorescente in Situ , Captura por Microdisección con Láser , Masculino , Datos de Secuencia Molecular , Proteínas Mutantes/metabolismo , Estadificación de Neoplasias , Polimorfismo de Nucleótido Simple/genética , Neoplasias de la Próstata/enzimología , Estructura Terciaria de Proteína , Análisis de Matrices Tisulares
11.
Blood ; 121(16): 3216-27, 2013 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-23327923

RESUMEN

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.


Asunto(s)
Antígenos CD34/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Células Eritroides/citología , Eritropoyesis , Eritropoyetina/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas Oncogénicas/metabolismo , Quinasas raf/metabolismo , Puntos de Control del Ciclo Celular , Proteínas de Ciclo Celular/genética , Proliferación Celular , Células Cultivadas , Proteínas de Unión al ADN/genética , Eritrocitos/citología , Eritrocitos/metabolismo , Células Eritroides/metabolismo , Células Precursoras Eritroides/citología , Células Precursoras Eritroides/metabolismo , Técnicas de Silenciamiento del Gen , Granulocitos/citología , Granulocitos/metabolismo , Humanos , Proteínas Oncogénicas/genética , ARN Mensajero/genética , Proteínas Son Of Sevenless/metabolismo , Regulación hacia Arriba
12.
Hum Vaccin Immunother ; 8(12): 1845-9, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22906933

RESUMEN

BACKGROUND: Guangdong province of China is HBV high endemicity and 1.6 million neonates are administrated 5 µg yeast recombinant anti-HBV vaccine each year. But few studies are performed to evaluate the immunogenicity and revaccination effect on non- and low- responders. METHODS: 2,199 children were administered intramuscularly with 5 µg vaccine at 0, 1 and 6 mo after birth. A 3 ml blood sample was drawn from each infant 1 mo after the third dose for determination of anti-HBs level. Three additional doses (10 µg each) were given to non- and low- responders. RESULTS: Of 1,814 subjects, 3.1% were non-responders (anti-HBs titer < 10 mIUml (-1) ) and 28.9% were low-responders (anti-HBs ≥ 10 mIUml (-1) and < 100 mIUml (-1) ). Low birth weight (LBW) was a risk factor for non- and low- responders (RR = 1.6, 95%CI = 1.2-2.0). After revaccination, of the 34 non-responders, 14.7% became low-responders and 85.3% became responders. Of the 74 low-responders, 21.6% remained at the same level and 78.4% shifted into responder category. CONCLUSIONS: Based on the lower responding rate after the primary immunization cycle and the higher responding rate after the additional cycle, measurement of anti-HBs level should be considered for people who had been immunized with three-dose 5 µg HB vaccine in Guangdong, especially for specific populations including LBW infants, healthcare workers, and patients with immunodeficiency disorders. An amount of 10 µg vaccine should be revaccinated to any non- and low- responders to provide adequate seroprotection.


Asunto(s)
Vacunas contra Hepatitis B/administración & dosificación , Vacunas contra Hepatitis B/inmunología , Hepatitis B/prevención & control , Inmunización Secundaria/métodos , China/epidemiología , Enfermedades Endémicas , Femenino , Hepatitis B/epidemiología , Anticuerpos contra la Hepatitis B/sangre , Humanos , Lactante , Recién Nacido , Inyecciones Intramusculares , Masculino , Resultado del Tratamiento , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología
13.
J Leukoc Biol ; 90(3): 529-38, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21653232

RESUMEN

Chemotaxis is fundamental to the directional migration of neutrophils toward endogenous and exogenous chemoattractants. Recent studies have demonstrated that ADF/cofilin superfamily members play important roles in reorganizing the actin cytoskeleton by disassembling actin filaments. GMFG, a novel ADF/cofilin superfamily protein that is expressed in inflammatory cells, has been implicated in regulating actin reorganization in microendothelial cells, but its function in neutrophils remains unclear. Here, we show that GMFG is an important regulator for cell migration and polarity in neutrophils. Knockdown of endogenous GMFG impaired fMLF- and IL-8 (CXCL8)-induced chemotaxis in dHL-60 cells. GMFG knockdown attenuated the formation of lamellipodia at the leading edge of cells exposed to fMLF or CXCL8, as well as the phosphorylation of p38 and PAK1/2 in response to fMLF or CXCL8. Live cell imaging revealed that GMFG was recruited to the leading edge of cells in response to fMLF, as well as CXCL8. Overexpression of GMFG enhanced phosphorylation of p38 but not of PAK1/2 in dHL-60 cells. In addition, we found that GMFG is associated with WAVE2. Taken together, our findings suggest that GMFG is a novel factor in regulating neutrophil chemotaxis by modulating actin cytoskeleton reorganization.


Asunto(s)
Movimiento Celular , Quimiotaxis , Factor de Maduración de la Glia/metabolismo , Neutrófilos/metabolismo , Seudópodos/metabolismo , Actinas/metabolismo , Adulto , Western Blotting , Adhesión Celular , Polaridad Celular , Proliferación Celular , Células Cultivadas , Citoesqueleto/metabolismo , Técnica del Anticuerpo Fluorescente , Factor de Maduración de la Glia/antagonistas & inhibidores , Factor de Maduración de la Glia/genética , Humanos , Inmunoprecipitación , Interleucina-8/farmacología , Neutrófilos/inmunología , Fosforilación , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Quinasas p21 Activadas/genética , Quinasas p21 Activadas/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
Carcinogenesis ; 32(7): 986-94, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21470957

RESUMEN

The human olfactomedin 4 gene (OLFM4) encodes an olfactomedin-related glycoprotein. OLFM4 is normally expressed in a limited number of tissues, including the prostate, but its biological functions in prostate are largely unknown. In this study, we found that OLFM4 messenger RNA was reduced or undetectable in prostate cancer tissues and prostate cancer cell lines. To study the effects of OLFM4 on prostate cancer progression, we transfected PC-3 prostate cancer cells with OLFM4 to establish OLFM4-expressing PC-3 cell clones. The OLFM4-expressing PC-3 cell clones were found to have decreased proliferation and invasiveness compared with vector-transfected control PC-3 cells in vitro. In addition, nude mice injected with OLFM4-expressing PC-3 cells demonstrated reduced tumor growth and bone invasion and metastasis compared with mice injected with vector-transfected control cells. Mechanistic studies revealed that OLFM4 may exhibit its anticancer effects through regulating cell autophagy by targeting cathepsin D, as OLFM4 reduced cathepsin D protein levels and enzymatic activity and attenuated cathepsin D-induced cancer cell proliferation. In addition, overexpression of OLFM4 abrogated stromal cell derived factor-1 (SDF-1)-induced PC-3 cell invasiveness in a Matrigel invasion assay, partially through blocking SDF-1-mediated AKT phosphorylation. Coimmunoprecipitation and immunofluorescence staining studies in OLFM4-expressing PC-3 cells demonstrated a direct interaction between OLFM4 and cathepsin D or SDF-1. Taken together, these results suggest that OLFM4 negatively interacts with cathepsin D and SDF-1 and inhibits prostate cancer growth and bone metastasis.


Asunto(s)
Catepsina D/metabolismo , División Celular , Quimiocina CXCL12/metabolismo , Factor Estimulante de Colonias de Granulocitos/fisiología , Metástasis de la Neoplasia , Neoplasias de la Próstata/patología , Electroforesis en Gel de Poliacrilamida , Citometría de Flujo , Humanos , Masculino , Neoplasias de la Próstata/metabolismo
15.
Blood ; 117(11): 3045-52, 2011 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-21220744

RESUMEN

The ß-hemoglobinopathies sickle cell disease and ß-thalassemia are among the most common human genetic disorders worldwide. Hemoglobin A2 (HbA2, α2δ2) and fetal hemoglobin (HbF, α2γ2) both inhibit the polymerization of hemoglobin S, which results in erythrocyte sickling. Expression of erythroid Kruppel-like factor (EKLF) and GATA1 is critical for transitioning hemoglobin from HbF to hemoglobin A (HbA, α2ß2) and HbA2. The lower levels of δ-globin expression compared with ß-globin expression seen in adulthood are likely due to the absence of an EKLF-binding motif in the δ-globin proximal promoter. In an effort to up-regulate δ-globin to increase HbA2 expression, we created a series of EKLF-GATA1 fusion constructs composed of the transactivation domain of EKLF and the DNA-binding domain of GATA1, and then tested their effects on hemoglobin expression. EKLF-GATA1 fusion proteins activated δ-, γ-, and ß-globin promoters in K562 cells, and significantly up-regulated δ- and γ-globin RNA transcript and protein expression in K562 and/or CD34(+) cells. The binding of EKLF-GATA1 fusion proteins at the GATA1 consensus site in the δ-globin promoter was confirmed by chromatin immunoprecipitation assay. Our studies demonstrate that EKLF-GATA1 fusion proteins can enhance δ-globin expression through interaction with the δ-globin promoter, and may represent a new genetic therapeutic approach to ß-hemoglobinopathies.


Asunto(s)
Células Eritroides/metabolismo , Factor de Transcripción GATA1/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Regulación hacia Arriba/genética , Globinas delta/genética , gamma-Globinas/genética , Antígenos CD34/metabolismo , Secuencia de Bases , Genes Reporteros , Humanos , Células K562 , Datos de Secuencia Molecular , Regiones Promotoras Genéticas/genética , Unión Proteica
16.
Blood ; 114(1): 187-94, 2009 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-19401563

RESUMEN

Increased fetal hemoglobin expression in adulthood is associated with acute stress erythropoiesis. However, the mechanisms underlying gamma-globin induction during the rapid expansion of adult erythroid progenitor cells have not been fully elucidated. Here, we examined COUP-TFII as a potential repressor of gamma-globin gene after stem cell factor (SCF) stimulation in cultured human adult erythroid progenitor cells. We found that COUP-TFII expression is suppressed by SCF through phosphorylation of serine/threonine phosphatase (PP2A) and correlated well with fetal hemoglobin induction. Furthermore, down-regulation of COUP-TFII expression with small interfering RNA (siRNA) significantly increases the gamma-globin expression during the erythroid maturation. Moreover, SCF-increased expression of NF-YA associated with redox regulator Ref-1 and cellular reducing condition enhances the effect of SCF on gamma-globin expression. Activation of Erk1/2 plays a critical role in SCF modulation of downstream transcriptional factor COUP-TFII, which is involved in the regulation of gamma-globin gene induction. Our data show that SCF stimulates Erk1/2 MAPK signaling pathway, which regulates the downstream repressor COUP-TFII by inhibiting serine/threonine phosphatase 2A activity, and that decreased COUP-TFII expression resulted in gamma-globin reactivation in adult erythropoiesis. These observations provide insight into the molecular pathways that regulate gamma-globin augmentation during stress erythropoiesis.


Asunto(s)
Factor de Transcripción COUP II/metabolismo , Factor de Células Madre/farmacología , gamma-Globinas/genética , Adulto , Secuencia de Bases , Sitios de Unión , Factor de Unión a CCAAT/genética , Factor de Unión a CCAAT/metabolismo , Células Cultivadas , Cartilla de ADN/genética , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Células Precursoras Eritroides/citología , Células Precursoras Eritroides/efectos de los fármacos , Células Precursoras Eritroides/metabolismo , Eritropoyesis/efectos de los fármacos , Eritropoyesis/genética , Eritropoyesis/fisiología , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Regiones Promotoras Genéticas , Proteína Fosfatasa 2/antagonistas & inhibidores , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes/farmacología , Estrés Fisiológico , Activación Transcripcional/efectos de los fármacos
17.
Zhongguo Yi Miao He Mian Yi ; 15(2): 152-8, 2009 Apr.
Artículo en Chino | MEDLINE | ID: mdl-20077662

RESUMEN

OBJECTIVE: To observe and compare the immunoreaction levels of revaccination with different hepatitis B vaccine in healthy population of different age groups. METHODS: Multi-stage random sampling method was used, 2 to 4 years old, 6 to 8 years old, 13 to 15 years old and 16 to 40 years old healthy people had been sampled in five towns according to different district. Screened those people's HBV serologic markers by ELISA, the person whose HBV serologic markers were all negative and had been vaccinated three doses of hepatitis B vaccine would be the revaccination recipients. Six kinds of recombinant Hepatitis B vaccine (yeast-derived or CHO cells) with 5 microg, 10 microg and 20 microg were used for the recipients on a 0-1-6 month schedule. RESULTS: Successful rates of revaccination and the median of antibody levels (mIU/ml) in the recipients were 94.34% and 226.53 in 5 microg yeast-derived vaccine group, 86.46% and 175.36 in 10 microg yeast-derived vaccine group, 97.39% and 331.44 in 10 microg CHO cells vaccine group, 91.30% and 439.01 in 10 microg Hansenula yeast-derived vaccine group, 99.20% and 386.66 in 20 microg CHO cells vaccine group, 89.04% and 372.97 in 20 microg yeast-derived vaccine group, 98.25% and 353.42 in 2 to 4 years old group, 96.80% and 320.31 in 6 to 8 years old group, 94.67% and 262.12 in 13 to 15 years old group, 87.76% and 305.24 in 16 to 40 years old group. CONCLUSION: Age, vaccine and dose were the most important factors to affect the immunoreaction levels of revaccination. There was an age-specific decline in immunogenicity, the immunoreaction levels of revaccination will be improved by changing vaccine.


Asunto(s)
Vacunas contra Hepatitis B/administración & dosificación , Virus de la Hepatitis B/inmunología , Hepatitis B/inmunología , Inmunización Secundaria , Adolescente , Adulto , Animales , Anticuerpos Antivirales/sangre , Células CHO , Niño , Preescolar , China , Cricetinae , Cricetulus , Femenino , Estado de Salud , Hepatitis B/prevención & control , Hepatitis B/virología , Vacunas contra Hepatitis B/inmunología , Humanos , Masculino , Adulto Joven
18.
Br J Haematol ; 141(2): 254-9, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18318767

RESUMEN

The hydroxycarbamide (HC)-inducible small guanosine triphosphate (GTP)-binding protein, secretion-associated and RAS-related (SAR) protein has recently been shown to play a pivotal role in HBG induction and erythroid maturation by causing cell apoptosis and G1/S-phase arrest. Our preliminary analysis indicated that HC inducibility is transcriptionally regulated by elements within the SAR1A promoter. This study aimed to assess whether polymorphisms in the SAR1A promoter are associated with differences Hb F levels or HC therapeutic responses among sickle cell disease (SCD) patients. We studied 386 individuals with SCD comprised of 269 adults treated with or without HC and 117 newborns with SCD identified from a newborn screening program. Three previously unknown single nucleotide polymorphisms (SNPs) in the upstream 5'UTR (-809 C>T, -502 G>T and -385 C>A) were significantly associated with the fetal haemoglobin (HbF) response in Hb SS patients treated with HC (P < 0.05). In addition, four SNPs (rs2310991, -809 C>T, -385 C>A and rs4282891) were significantly associated with the change in absolute HbF after 2 years of treatment with HC. These data suggest that variation within SAR1A regulatory elements might contribute to inter-individual differences in regulation of HbF expression and patient responses to HC in SCD.


Asunto(s)
Anemia de Células Falciformes/tratamiento farmacológico , Antidrepanocíticos/uso terapéutico , Hemoglobina Fetal/metabolismo , Hidroxiurea/uso terapéutico , Proteínas de Unión al GTP Monoméricas/genética , Adulto , Anemia de Células Falciformes/sangre , Anemia de Células Falciformes/genética , Humanos , Recién Nacido , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas/genética
19.
Clin Cancer Res ; 14(4): 1041-9, 2008 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-18281536

RESUMEN

PURPOSE: hGC-1 (human granulocyte colony-stimulating factor-stimulated clone 1) is a gastrointestinal protein that is a member of the olfactomedin glycoprotein family. Its biological function remains poorly understood. Aberrant expression of hGC-1 in some human carcinomas has been recently reported. The purpose of this study was to examine hGC-1 expression in colon carcinoma and explore the relationship between hGC-1 expression and the clinicopathologic features of patients with colon cancer. EXPERIMENTAL DESIGN: The expression of hGC-1 in colon adenocarcinoma tissues was examined by dot-blot analysis, in situ hybridization, and immunohistochemistry. The association of hGC-1 expression pattern with patient differentiation grade, tumor stage, metastasis, and survival were examined. To further investigate the involvement of hGC-1 in colon cancer progression, human colon carcinoma (HT-29) cells overexpressing hGC-1 were established and cell proliferation, adhesion, and migration were studied. RESULTS: Compared with normal colon mucosa, the up-regulation of hGC-1 was more frequently detected in more differentiated colon cancers, whereas down-regulation or no expression was associated with poorly differentiated colon cancers. Interestingly, hGC-1 down-regulation was also found in late tumor-node-metastasis stage, metastasis, and in patients with shorter survival. The morphology and cortical actin distribution of HT-29 cells were altered by hGC-1 overexpression. However, this did not change cell proliferation, but decreased cell adhesion and migration. CONCLUSION: Our findings indicate that hGC-1 is involved in colon cancer adhesion and metastasis, and that hGC-1 may be a useful marker for tumor differentiation and progression of human colon carcinoma.


Asunto(s)
Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Factor Estimulante de Colonias de Granulocitos/biosíntesis , Adenocarcinoma/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/análisis , Adhesión Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Neoplasias del Colon/genética , Metilación de ADN , Análisis Mutacional de ADN , Progresión de la Enfermedad , Femenino , Expresión Génica , Factor Estimulante de Colonias de Granulocitos/genética , Células HT29 , Humanos , Immunoblotting , Inmunohistoquímica , Hibridación in Situ , Estimación de Kaplan-Meier , Masculino , Microscopía Confocal , Persona de Mediana Edad , Regiones Promotoras Genéticas , ARN Mensajero/análisis
20.
Artículo en Chino | MEDLINE | ID: mdl-19544628

RESUMEN

OBJECTIVE: To develop pathogenic surveillance on measles and to effectively isolate measles virus. To know the genetic characterizations and molecular epidemiology of wildtype measles viruses from 2005 to 2007 in Guangdong Province, and provide the scientific basis for measles control and eradication. METHODS: Vero/Slam cell line were used, measles viruses were isolated from throat swabs or urine specimens collected from uspected measles patients in outbreaks and sporadic patients. A 450 nucleotides fragment of the C-terminal of the nucleoprotein (N) gene was amplified and by RT-PCR and subjected to sequence and phylogenetic analysis using Bio-Edit software. RESULTS: 82 wild-type measels virus were obtained from 377 throat swabs and urine specimens from 2005 to 2007 in Guangdong Province measles lab. The measles isolation rate was 23.58% in 2005, 17.11% in 2006, 39.13% in 2007. The succeed rate of virus isolation is related to the quality of specimens collected and the days after rash occurrence. CONCLUSIONS: We have grasped the technicalability of measles virus isolation and confirm action, and got higher isolation ratio. The wild-type measles virus isolated from Guangdong Province is of H1 genotype from 2005 to 2007, which is the same as the dominant genotype circulation.


Asunto(s)
Virus del Sarampión/aislamiento & purificación , Sarampión/diagnóstico , Vigilancia de la Población/métodos , China/epidemiología , Genotipo , Humanos , Sarampión/epidemiología , Sarampión/genética , Virus del Sarampión/clasificación , Virus del Sarampión/genética , Epidemiología Molecular , Datos de Secuencia Molecular , Filogenia , Plasma/virología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Orina/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...