Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Thorac Cancer ; 12(16): 2258-2264, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34236145

RESUMEN

BACKGROUND: Circular RNAs (circRNAs) had been identified as a non-coding RNA associated with many types of cancer in recent years. However, the involvement of hsa_circ_0008274 in lung adenocarcinoma (LUAD) has not been explored. The aim of our research is to explore the biological mechanism and function of hsa_circ_0008274 in LUAD. METHODS: The hsa_circ_0008274, miR-578, and high mobility group AT-Hook 2 (HMGA2) mRNA expression levels were detected via qRT-PCR. Cell Counting Kit-8 (CCK-8) Transwell assay and wound healing assay were performed to measure the cell proliferation, invasion, and migration ability. Luciferase reporter and Western blotting experiments were performed to characterize the competing endogenous RNA (ceRNA) mechanism of hsa_circ_0008274. RESULTS: Our findings determined that the expression of hsa_circ_0008274 in LUAD was significantly decreased. Cell experiments showed that overexpressed hsa_circ_0008274 could reduce the proliferation and invasion ability of LUAD cells. Moreover, miRNA-578 could identify as a miRNA sponge of hsa_circ_0008274. Overexpressed hsa_circ_0008274 reduced the proliferation and invasion of LUAD cells caused by miR-578 mimics. Increasing the expression of miR-578 can aggravate the proliferation and invasion of LUAD cells and block the inhibition of proliferation and invasion of LUAD cells mediated by overexpressed hsa_circ_0008274. Subsequent data indicate that HMGA2 of the tumor-promoting gene is the target gene of miR-578. The upregulation of HMGA2 partially reversed the tumor inhibitory effect of LUAD cells induced by overexpressed hsa_circ_0008274 or miR-578 mimics. CONCLUSIONS: In summary, our data show that the overexpression of hsa_circ_0008274 repressed the proliferation and invasion of LUAD through downregulating miR-578 and activating HMGA2.


Asunto(s)
Adenocarcinoma del Pulmón/genética , Proteína HMGA2/fisiología , Neoplasias Pulmonares/genética , MicroARNs/fisiología , ARN Circular/fisiología , Células A549 , Proliferación Celular/genética , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Humanos
2.
J Orthop Surg Res ; 15(1): 374, 2020 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-32883329

RESUMEN

BACKGROUND: Osteosarcoma (OS) is a common severe illness globally. Lupeol has been reported to participate in the pathophysiologic properties of various cancers, including OS. This study aimed to explore the effects of lupeol on proliferation, invasion, and apoptosis on OS cells and the underlying mechanism. METHODS: The cell viability of OS cells was determined by 3-(4, 5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) assay. The expression levels of miR-212-3p and high-mobility group AT-hook 2 (HMGA2) were detected by quantitative real-time polymerase chain reaction (qRT-PCR) in OS cells. The cell apoptosis and invasion were detected by flow cytometry and transwell invasion assays, respectively. The functional target of miR-212-3p was predicted by online software and confirmed by luciferase reporter assay. The protein level of HMGA2 was measured by western blot analysis. RESULTS: Lupeol suppressed cell viability and invasion, and promoted apoptosis by upregulating the expression of miR-212-3p in OS cells. Knockdown of miR-212-3p restored the anti-tumor effect of lupeol. Interestingly, miR-212-3p directly targeted HMGA2 and suppressed its expression. Moreover, HMGA2 reversed the inhibited impact on viability and invasion, and the promoted effect on apoptosis induced by upregulation of miR-212-3p. Also, lupeol administration exerts its anti-tumor effect by overexpression of miR-212-3p to suppress the expression of HMGA2 in OS cells. CONCLUSION: Lupeol inhibited OS progression by modulating the miR-212-3p/HMGA2 axis in vitro.


Asunto(s)
Neoplasias Óseas/genética , Neoplasias Óseas/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteína HMGA2/genética , Proteína HMGA2/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Osteosarcoma/genética , Osteosarcoma/patología , Triterpenos Pentacíclicos/farmacología , Regulación hacia Arriba/efectos de los fármacos , Antineoplásicos , Apoptosis/efectos de los fármacos , Apoptosis/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Progresión de la Enfermedad , Proteína HMGA2/fisiología , Humanos , Invasividad Neoplásica/genética , Células Tumorales Cultivadas
3.
Biomed Pharmacother ; 121: 109622, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31707340

RESUMEN

BACKGROUND: Previous study has demonstrated that long noncoding RNA cyclin-dependent kinase inhibitor 2B antisense RNA 1 (CDKN2B-AS1) was abnormally expressed in diabetic nephropathy (DN). However, the underlying mechanism that allows CDKN2B-AS1 in the progression of DN remains to be further elucidated. METHODS: Peripheral blood cells of 24 diabetes patients with DN and 20 without DN were collected. Human glomerular mesangial cells (HGMC) were cultured in high glucose or low glucose medium. The expression levels of CDKN2B-AS1, microRNA (miR)-424-5p and high mobility group AT hook 2 (HMGA2) were detected by quantitative real-time polymerase chain reaction or western blot. The target association between miR-424-5p and CDKN2B-AS1 or HMGA2 was confirmed by dual-luciferase reporter and RNA immunoprecipitation assays. Cell proliferation, extracellular matrix (ECM) accumulation and phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling were investigated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT) and western blot, respectively. RESULTS: CDKN2B-AS1 expression was up-regulated and miR-424-5p level was down-regulated in peripheral blood of DN patients and high glucose-treated HGMC cells. CDKN2B-AS1 was validated as a sponge of miR-424-5p. Silence of CDKN2B-AS1 repressed proliferation and ECM accumulation by increasing miR-424-5p. HMGA2 was a target of miR-424-5p and miR-424-5p overexpression inhibited proliferation, ECM accumulation and PI3K/AKT pathway by targeting HMGA2. Moreover, knockdown of CDKN2B-AS1 inhibited HMGA2 expression and PI3K/AKT pathway by increasing miR-424-5p. CONCLUSION: Knockdown of CDKN2B-AS1 suppressed proliferation, ECM accumulation and PI3K/AKT signaling by increasing miR-424-5p and decreasing HMGA2 in high glucose-treated HMGC cells.


Asunto(s)
Nefropatías Diabéticas/etiología , Matriz Extracelular/metabolismo , Proteína HMGA2/fisiología , Células Mesangiales/fisiología , MicroARNs/fisiología , ARN Largo no Codificante/fisiología , Proliferación Celular , Células Cultivadas , Nefropatías Diabéticas/metabolismo , Humanos , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología
4.
Gene ; 706: 131-139, 2019 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-31055021

RESUMEN

The high mobility group A2 (HMGA2; also called HMGI-C) gene is an architectural transcription factor that belonging to the high mobility group AT-hook (HMGA) gene family. HMGA2 is aberrantly regulated in several human tumors. Over-expression of HMGA2 is correlated with a higher risk of metastasis and an unfavorable prognosis in patients with cancer. We performed a meta-analysis to determine the clinic-pathological and prognostic value of HMGA2 overexpression in different human tumors. A comprehensive literature search was performed using PubMed, Embase, Cochrane Library, Scopus, MEDLINE, Google Scholar and ISI Web of Science. Hazard ratios (HRs)/odds ratios (ORs) and their 95% confidence intervals (CIs) were used to assess the strength of the association between HMGA2 expression and overall survival (OS)/progression free survival (PFS)/disease free survival (DFS). A total of 5319 patients with 19 different types of cancer from 35 articles were evaluated. Pooled data analysis indicated that increased HMGA2 expression in cancer patients predicted a poor OS (HR = 1.70; 95% CI = 1.6-1.81; P < 0.001; fixed-effect model). In subgroup analyses, high HMGA2 expression was particularly associated with poor OS in individuals with gastrointestinal (GI) cancer (HR = 1.89, 95% CI: 1.83-1.96; fixed-effect model) and HNSCC cancer (HR-1.78, 95%CI: 1.44-2.21; fixed-effect model). Over-expression of HMGA2 was associated with vascular invasion (OR = 0.16, 95% CI = 0.05-0.49; P = 0.001) and lymphatic invasion (OR = 1.89, 95% CI = 1.06-3.38; P = 0.032). Further studies should be conducted to validate the prognostic value of HMGA2 for patients with GI cancers.


Asunto(s)
Proteína HMGA2/genética , Neoplasias/genética , Biomarcadores de Tumor/metabolismo , Supervivencia sin Enfermedad , Femenino , Proteína HMGA2/fisiología , Proteínas del Grupo de Alta Movilidad/genética , Humanos , Masculino , Pronóstico , Modelos de Riesgos Proporcionales , Análisis de Supervivencia
5.
Biochem Biophys Res Commun ; 509(4): 1008-1014, 2019 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-30654938

RESUMEN

Tooth formation is accomplished under strict genetic programs. Although patients with chromosome 12q14 aberration shows tooth phenotype including the size and eruption timing with bone growth anomaly, its etiology is uncertain. Here, we examined expression of Hmga2, which is encoded at chromosome 12q14, in mouse tooth germs and analyzed the involvement in lower first molar (M1) and mandibular bone development. Hmga2 expression was immunohistochemically detected at enamel organ and the surrounding mesenchyme of the M1 germs. The expression was dynamically changed with gestation and rapidly decreased in postnatal mice. In Hmga2-/- mice, the M1 germs and crowns were diminished in size, and formation and eruption of molars were delayed with mandibular bone growth retardation. Hmga2 cDNA or siRNA transfection showed that Hmga2 transcriptionally up-regulates expression of stem cell factors, Sox2 and Nanog. They were co-localized with Hmga2 in the germs, but differentially distributed at enamel organ and mesenchyme in Hmga2-/- mice. These results demonstrate that Hmga2 expressed in tooth germs regulates the growth, sizing and eruption and stem cell factor expression in different compartment of the germ and associates with mandibular bone growth. Although future studies are needed, the present study demonstrates HMGA2 regulation of tooth genesis with skeletal development.


Asunto(s)
Proteína HMGA2/fisiología , Proteína Homeótica Nanog/metabolismo , Factores de Transcripción SOXB1/metabolismo , Animales , Regulación del Desarrollo de la Expresión Génica , Proteína HMGA2/análisis , Proteína HMGA2/metabolismo , Inmunohistoquímica , Mandíbula/crecimiento & desarrollo , Ratones , Diente Molar/crecimiento & desarrollo , Odontogénesis/efectos de los fármacos
6.
Oncol Res ; 27(2): 173-182, 2019 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-29523225

RESUMEN

The long noncoding RNA (lncRNA) H19 has been described to participate in the metastasis of various tumors. Nevertheless, whether H19 promotes or impedes tongue squamous cell carcinoma (TSCC) cell migration and invasion remains controversial. Here we found that the expression of H19 was elevated in TSCC tissues compared with adjacent normal tissues. Moreover, we demonstrated that the expression of H19 was higher in metastasized tumors compared with unmetastasized tumors. Consistently, TSCC cells express higher levels of H19 than human squamous cells. Subsequently, depletion of H19 impaired the migration and invasion abilities of TSCC cells. Mechanistically, we demonstrated that H19 functions as a competing endogenous RNA (ceRNA) to sponge miRNA let-7a, leading to an increase in a let-7a target, the key regulator of tumor metastasis HMGA2, which is enriched in TSCC tissues and cell lines. Intriguingly, inhibition of let-7a significantly rescued the short hairpin H19 (shH19)-induced decrease in TSCC migration and invasion. These findings revealed that the H19/let-7a/HMGA2/EMT axis plays a critical role in the regulation of TSCC migration and invasion, which may provide a new therapeutic target for TSCC.


Asunto(s)
MicroARNs/fisiología , ARN Largo no Codificante/fisiología , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Neoplasias de la Lengua/patología , Adulto , Línea Celular Tumoral , Movimiento Celular , Transición Epitelial-Mesenquimal , Femenino , Proteína HMGA2/análisis , Proteína HMGA2/fisiología , Humanos , Masculino , MicroARNs/antagonistas & inhibidores , Persona de Mediana Edad , Invasividad Neoplásica
7.
Breast Cancer Res Treat ; 172(3): 577-586, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30220054

RESUMEN

PURPOSE: Metaplastic breast carcinomas are an aggressive subtype of triple-negative breast cancer (TNBC) in which part or all of the adenocarcinoma transforms into a non-glandular component (e.g., spindled, squamous, or heterologous). We discovered that mammary-specific Ccn6/Wisp3 knockout mice develop mammary carcinomas with spindle and squamous differentiation that share upregulation of the oncofetal proteins IGF2BP2 (IMP2) and HMGA2 with human metaplastic carcinomas. Here, we investigated the functional relationship between CCN6, IGF2BP2, and HMGA2 proteins in vitro and in vivo, and their expression in human tissue samples. METHODS: MMTV-cre;Ccn6fl/fl tumors and spindle TNBC cell lines were treated with recombinant CCN6 protein or vehicle. IGF2BP2 was downregulated using shRNAs in HME cells with stable CCN6 shRNA knockdown, and subjected to invasion and adhesion assays. Thirty-one human metaplastic carcinomas were arrayed in a tissue microarray (TMA) and immunostained for CCN6, IGF2BP2, and HMGA2. RESULTS: CCN6 regulates IGF2BP2 and HMGA2 protein expression in MMTV-cre;Ccn6fl/fl tumors, in MDA-MB-231 and - 468, and in HME cells. CCN6 recombinant protein reduced IGF2BP2 and HMGA2 protein expression, and decreased growth of MMTV-cre;Ccn6fl/fl tumors in vivo. IGF2BP2 shRNA knockdown was sufficient to reverse the invasive abilities conferred by CCN6 knockdown in HME cells. Analyses of the TCGA Breast Cancer Cohort (n = 1238) showed that IGF2BP2 and HMGA2 are significantly upregulated in metaplastic carcinoma compared to other breast cancer subtypes. In clinical samples, low CCN6 is frequent in tumors with high IGF2BP2/HMGA2 with spindle and squamous differentiation. CONCLUSIONS: These data shed light into the pathogenesis of metaplastic carcinoma and demonstrate a novel CCN6/IGF2BP2/HMGA2 oncogenic pathway with biomarker and therapeutic implications.


Asunto(s)
Proteínas CCN de Señalización Intercelular/fisiología , Proteína HMGA2/fisiología , Proteínas de Unión al ARN/fisiología , Transducción de Señal/fisiología , Neoplasias de la Mama Triple Negativas/patología , Animales , Adhesión Celular , Línea Celular Tumoral , Femenino , Proteína HMGA2/análisis , Humanos , Ratones , Persona de Mediana Edad , Invasividad Neoplásica , Metástasis de la Neoplasia , Proteínas de Unión al ARN/análisis , Neoplasias de la Mama Triple Negativas/terapia
8.
Oncol Rep ; 37(1): 185-192, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27878307

RESUMEN

High expression of high mobility group protein A2 (HMGA2) is correlated with the invasiveness of gastric cancer and is an independent prognostic factor. The reason may be that HMGA2 promotes epithelial-mesenchymal transition (EMT) and the acquisition of tumor stem cell properties, yet the mechanism remains unclear. In this study, immunohistochemistry and western blot analysis revealed that the expression of HMGA2 and Twist-related protein 1 (TWIST1) in gastric carcinoma tissues was higher than that in the peritumoral tissues and that the expression levels of these two proteins were positively correlated. The protein expression levels of HMGA2 and TWIST1 were high in the poorly differentiated gastric cancer MKN-45 cells and were low in the moderately differentiated SGC-7901 cells. TWIST1 was inhibited after HMGA2 interference and was significantly increased after overexpression of HMGA2. Luciferase experiments showed that TWIST1 was a direct downstream target gene of HMGA2. The simultaneous interference of HMGA2 expression and the overexpression of TWIST1 in MKN-45 cells reversed the inhibitory effect of HMGA2 interference on the invasion and migration of gastric cancer cells, EMT and the expression of stemness markers. However, the simultaneous overexpression of HMGA2 and the interference of TWIST1 expression in the SGC-7901 cells reversed the promoter effect of HMGA2 overexpression on the invasiveness and migration of gastric cancer cells, EMT and the expression of stemness markers. In addition, animal experiments showed that TWIST1 overexpression reversed the inhibition of HMGA2 interference on the metastasis of MKN-45 cells. Therefore, HMGA2 regulates the EMT of gastric cancer cells and the acquisition of tumor stem cell properties through direct regulation of the downstream target gene TWIST1.


Asunto(s)
Transición Epitelial-Mesenquimal/genética , Proteína HMGA2/fisiología , Células Madre Neoplásicas/metabolismo , Proteínas Nucleares/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Proteína 1 Relacionada con Twist/genética , Animales , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Células Hep G2 , Humanos , Ratones , Ratones Endogámicos BALB C , Células Madre Neoplásicas/patología
9.
Nucleic Acids Res ; 44(22): e162, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27587582

RESUMEN

HMGA2 is an important chromatin factor that interacts with DNA via three AT-hook domains, thereby regulating chromatin architecture and transcription during embryonic and fetal development. The protein is absent from differentiated somatic cells, but aberrantly re-expressed in most aggressive human neoplasias where it is causally linked to cell transformation and metastasis. DNA-binding also enables HMGA2 to protect cancer cells from DNA-damaging agents. HMGA2 therefore is considered to be a prime drug target for many aggressive malignancies. Here, we have developed a broadly applicable cell-based reporter system which can identify HMGA2 antagonists targeting functionally important protein domains, as validated with the known AT-hook competitor netropsin. In addition, high-throughput screening can uncover functional links between HMGA2 and cellular factors important for cell transformation. This is demonstrated with the discovery that HMGA2 potentiates the clinically important topoisomerase I inhibitor irinotecan/SN-38 in trapping the enzyme in covalent DNA-complexes, thereby attenuating transcription.


Asunto(s)
Camptotecina/análogos & derivados , ADN-Topoisomerasas de Tipo I/fisiología , Proteína HMGA2/fisiología , Inhibidores de Topoisomerasa I/farmacología , Secuencia de Aminoácidos , Camptotecina/farmacología , Diferenciación Celular/efectos de los fármacos , Estabilidad de Enzimas , Genes Reporteros , Células HEK293 , Células HeLa , Ensayos Analíticos de Alto Rendimiento , Humanos , Irinotecán , Luciferasas de Renilla/biosíntesis , Luciferasas de Renilla/genética , Netropsina/farmacología , Regiones Promotoras Genéticas , Transcripción Genética/efectos de los fármacos , Activación Transcripcional
10.
Hum Reprod ; 31(6): 1300-14, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27083540

RESUMEN

STUDY QUESTION: What are the functional characteristics and transcriptional regulators of human trophoblast progenitor cells (TBPCs)? SUMMARY ANSWER: TBPC lines established from the human smooth chorion by cell sorting for integrin α4 expressed markers of stemness and trophoblast (TB) stage-specific antigens, invaded Matrigel substrates and contributed to the cytotrophoblasts (CTBs) layer of smooth chorion explants with high-mobility group protein HMGI-C (HMGA2) and transcription factor GATA-4 (GATA4) controlling their progenitor state and TB identity. WHAT IS KNOWN ALREADY: Previously, we reported the derivation of TBPC lines by trypsinization of colonies that formed in cultures of chorionic mesenchyme cells that were treated with an activin nodal inhibitor. Microarray analyses showed that, among integrins, α4 was most highly expressed, and identified HMGA2 and GATA4 as potential transcriptional regulators. STUDY DESIGN, SIZE, DURATION: The aim of this study was to streamline TBPC derivation across gestation. High-cell surface expression of integrin α4 enabled the use of a fluorescence-activated cell sorter (FACS) approach for TBPC isolation from the human smooth chorion (n = 6 lines). To confirm their TBPC identity, we profiled their expression of stemness and TB markers, and growth factor receptors. At a functional level, we assayed their invasive capacity (n = 3) and tropism for the CTB layer of the smooth chorion (n = 3). At a molecular level, we studied the roles of HMGA2 and GATA4. PARTICIPANTS/MATERIALS, SETTINGS, METHODS: Cells were enzymatically disassociated from the human smooth chorion across gestation. FACS was used to isolate the integrin α4-positive population. In total, we established six TBPC lines, two per trimester. Their identity was determined by immunolocalization of a suite of antigens. Function was assessed via Matrigel invasion and co-culture with explants of the human smooth chorion. An siRNA approach was used to down-regulate HMGA2 and GATA4 expression and the results were confirmed by immunoblotting and quantitative reverse transcription-polymerase chain reaction (qRT-PCR) analyses. The endpoints analyzed included proliferation, as determined by 5-bromo-2'-deoxyuridine (BrDU) incorporation, and the expression of stage-specific antigens and hormones, as determined by qRT-PCR and immunostaining approaches. MAIN RESULTS AND THE ROLE OF CHANCE: As with the original cell lines, the progenitors expressed a combination of human embryonic stem cell and TB markers. Upon differentiation, they primarily formed CTBs, which were capable of Matrigel invasion. Co-culture of the cells with smooth chorion explants enabled their migration through the mesenchyme after which they intercalated within the chorionic CTB layer. Down-regulation of HMGA2 showed that this DNA-binding protein governed their self-renewal. Both HMGA2 and GATA4 had pleitropic effects on the cells' progenitor state and TB identity. LIMITATIONS, REASONS FOR CAUTION: This study supported our hypothesis that TBPCs from the chorionic mesenchyme can contribute to the subpopulation of CTBs that reside in the smooth chorion. In the absence of in vivo data, which is difficult to obtain in humans, the results have the limitations common to all in vitro studies. WIDER IMPLICATIONS OF THE FINDINGS: The accepted view is that progenitors reside among the villous CTB subpopulation. Here, we show that TBPCs also reside in the mesenchymal layer of the smooth chorion throughout gestation. We theorize that they can contribute to the CTB layer in this region. This phenomenon may be particularly important in pathological situations when CTBs of the smooth chorion might provide a functional reserve for CTBs of the placenta proper. STUDY FUNDING/COMPETING INTERESTS: Research reported in this publication was supported by the Eunice Kennedy Shriver National Institute of Child Health and Human Development of the National Institutes of Health under award P50HD055764. O.G., N.L., K.O., A.P., T.G.-G., M.K., A.B., M.G. have nothing to disclose. S.J.F. received licensing fees and royalties from SeraCare Life Sciences for trisomic TBPC lines that were derived according to the methods described in this manuscript. TRIAL REGISTRATION NUMBER: N/A.


Asunto(s)
Factor de Transcripción GATA4/fisiología , Integrina alfa4/metabolismo , Trofoblastos/metabolismo , Diferenciación Celular , Línea Celular , Corion/citología , Corion/metabolismo , Técnicas de Cocultivo , Citometría de Flujo , Factor de Transcripción GATA4/genética , Factor de Transcripción GATA4/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteína HMGA2/genética , Proteína HMGA2/metabolismo , Proteína HMGA2/fisiología , Humanos , Integrina alfa4/genética , Elementos Reguladores de la Transcripción
11.
Dev Biol ; 411(1): 25-37, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26806704

RESUMEN

HMGA proteins are small nuclear proteins that bind DNA by conserved AT-hook motifs, modify chromatin architecture and assist in gene expression. Two HMGAs (HMGA1 and HMGA2), encoded by distinct genes, exist in mammals and are highly expressed during embryogenesis or reactivated in tumour progression. We here addressed the in vivo role of Xenopus hmga2 in the neural crest cells (NCCs). We show that hmga2 is required for normal NCC specification and development. hmga2 knockdown leads to severe disruption of major skeletal derivatives of anterior NCCs. We show that, within the NCC genetic network, hmga2 acts downstream of msx1, and is required for msx1, pax3 and snail2 activities, thus participating at different levels of the network. Because of hmga2 early effects in NCC specification, the subsequent epithelial-mesenchymal transition (EMT) and migration of NCCs towards the branchial pouches are also compromised. Strictly paralleling results on embryos, interfering with Hmga2 in a breast cancer cell model for EMT leads to molecular effects largely consistent with those observed on NCCs. These data indicate that Hmga2 is recruited in key molecular events that are shared by both NCCs and tumour cells.


Asunto(s)
Diferenciación Celular/genética , Transición Epitelial-Mesenquimal/genética , Proteína HMGA2/fisiología , Cresta Neural/embriología , Proteínas de Xenopus/fisiología , Xenopus laevis/embriología , Animales , Línea Celular Tumoral , Movimiento Celular/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes/genética , Proteína HMGA2/genética , Factor de Transcripción MSX1/genética , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Morfolinos/genética , Cresta Neural/citología , Factor de Transcripción PAX3 , Factores de Transcripción Paired Box/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Factores de Transcripción/genética , Factor de Crecimiento Transformador beta/metabolismo , Proteínas de Xenopus/genética
12.
Oncogene ; 35(12): 1529-40, 2016 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-26165842

RESUMEN

As the majority of patients with basal-like breast carcinoma present with invasive, metastatic disease that do not respond to available therapies, it is essential to identify new therapeutic targets that impact invasion and metastasis. Protease-activated receptor 1 (PAR1), a G-protein coupled receptor has been shown to act as an oncogene, but underlying mechanisms are not well understood. Here, we show that ectopic expression of functionally active PAR1 in MCF-7 cells induced a hormone-refractory, invasive phenotype representative of advanced basal-like breast carcinoma that readily formed metastatic lesions in lungs of mice. PAR1 was found to globally upregulate mesenchymal markers, including vimentin, a direct target of PAR1, and downregulate the epithelial markers including E-cadherin, as well as estrogen receptor. In contrast, non-signaling PAR1 mutant receptor did not lead to an invasive, hormone refractory phenotype. PAR1 expression increased spheroid formation and the level of stemness markers and self-renewal capacity in human breast cancer cells. We identified HMGA2 (high mobility group A2) as an important regulator of PAR1-mediated invasion. Inhibition of PAR1 signaling suppresses HMGA2-driven invasion in breast cancer cells. HMGA2 gene and protein are highly expressed in metastatic breast cancer cells. Overall, our results show that PAR1/HMGA2 pathway may present a novel therapeutic target.


Asunto(s)
Neoplasias de la Mama/patología , Proteína HMGA2/fisiología , Metástasis de la Neoplasia/fisiopatología , Receptor PAR-1/fisiología , Femenino , Humanos , Células MCF-7 , Fenotipo , Vimentina/metabolismo
13.
Dev Biol ; 410(1): 70-85, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26698218

RESUMEN

One of the well established aspects of the development of the central nervous system (CNS) is that neurogenesis precedes gliogenesis. However, the mechanism underlying this temporal switch between the two distinct lineages is poorly understood. It is thought that let-7, a heterochronic miRNA, may help neural stem cells (NSCs) choose between the neuronal and glial lineages. Here, we have tested the premise in the retina, a simple and accessible CNS model, where neurogliogenic decision takes place postnatally during late histogenesis. A positive correlation was observed between the temporal induction of let-7 expression and differentiation of late born neurons and Müller glia (MG), the sole glia generated by retinal progenitor cells (RPCs). Examination of let-7's involvement in late histogenesis by the perturbation of function approaches revealed that let-7 facilitated differentiation of both neurons and MG, without preference to a particular lineage. We demonstrate that let-7's positive influence on neuronal and MG differentiation is likely achieved by inhibiting the expression of Hmga2, the DNA architecture protein involved in the self-renewal of neural progenitors. Thus, our observations suggest that let-7 promotes differentiation, regardless of the neuronal or glial lineage, shifting the balance from RPCs' maintenance to their differentiation in the developing retina.


Asunto(s)
Proteína HMGA2/fisiología , MicroARNs/fisiología , Neurogénesis , Neuroglía/fisiología , Retina/embriología , Animales , Diferenciación Celular , Ratas , Ratas Sprague-Dawley
14.
Cancer Res ; 75(19): 4063-73, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26238785

RESUMEN

Triple-negative breast cancer (TNBC) patients have the highest risk of recurrence and metastasis. Because they cannot be treated with targeted therapies, and many do not respond to chemotherapy, they represent a clinically underserved group. TNBC is characterized by reduced expression of metastasis suppressors such as Raf kinase inhibitory protein (RKIP), which inhibits tumor invasiveness. Mechanisms by which metastasis suppressors alter tumor cells are well characterized; however, their ability to regulate the tumor microenvironment and the importance of such regulation to metastasis suppression are incompletely understood. Here, we use species-specific RNA sequencing to show that RKIP expression in tumors markedly reduces the number and metastatic potential of infiltrating tumor-associated macrophages (TAM). TAMs isolated from nonmetastatic RKIP(+) tumors, relative to metastatic RKIP(-) tumors, exhibit a reduced ability to drive tumor cell invasion and decreased secretion of prometastatic factors, including PRGN, and shed TNFR2. RKIP regulates TAM recruitment by blocking HMGA2, resulting in reduced expression of numerous macrophage chemotactic factors, including CCL5. CCL5 overexpression in RKIP(+) tumors restores recruitment of prometastatic TAMs and intravasation, whereas treatment with the CCL5 receptor antagonist Maraviroc reduces TAM infiltration. These results highlight the importance of RKIP as a regulator of TAM recruitment through chemokines such as CCL5. The clinical significance of these interactions is underscored by our demonstration that a signature comprised of RKIP signaling and prometastatic TAM factors strikingly separates TNBC patients based on survival outcome. Collectively, our findings identify TAMs as a previously unsuspected mechanism by which the metastasis-suppressor RKIP regulates tumor invasiveness, and further suggest that TNBC patients with decreased RKIP activity and increased TAM infiltration may respond to macrophage-based therapeutics.


Asunto(s)
Quimiocinas/fisiología , Quimiotaxis , Macrófagos/inmunología , Neoplasias Mamarias Experimentales/inmunología , Metástasis de la Neoplasia/inmunología , Proteínas de Neoplasias/fisiología , Proteínas de Unión a Fosfatidiletanolamina/fisiología , Neoplasias de la Mama Triple Negativas/inmunología , Microambiente Tumoral/inmunología , Animales , Línea Celular Tumoral/trasplante , Quimiocina CCL5/biosíntesis , Quimiocina CCL5/genética , Quimiocina CCL5/fisiología , Ciclohexanos/farmacología , Ciclohexanos/uso terapéutico , Supervivencia sin Enfermedad , Femenino , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Proteína HMGA2/fisiología , Xenoinjertos/inmunología , Humanos , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Maraviroc , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética , Receptores CCR5/efectos de los fármacos , Análisis de Secuencia de ARN , Triazoles/farmacología , Triazoles/uso terapéutico , Neoplasias de la Mama Triple Negativas/mortalidad
15.
Annu Rev Cell Dev Biol ; 30: 535-60, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25062362

RESUMEN

Although most modern dog breeds are less than 200 years old, the symbiosis between man and dog is ancient. Since prehistoric times, repeated selection events have transformed the wolf into man's guardians, laborers, athletes, and companions. The rapid transformation from pack predator to loyal companion is a feat that is arguably unique among domesticated animals. How this transformation came to pass remained a biological mystery until recently: Within the past decade, the deployment of genomic approaches to study population structure, detect signatures of selection, and identify genetic variants that underlie canine phenotypes is ushering into focus novel biological mechanisms that make dogs remarkable. Ironically, the very practices responsible for breed formation also spurned morbidity; today, many diseases are correlated with breed identity. In this review, we discuss man's best friend in the context of a genetic model to understand paradigms of heritable phenotypes, both desirable and disadvantageous.


Asunto(s)
Perros/genética , Genoma , Animales , Tamaño Corporal/genética , Neoplasias Óseas/genética , Neoplasias Óseas/veterinaria , Cruzamiento , Mapeo Cromosómico , Modelos Animales de Enfermedad , Enfermedades de los Perros/genética , Perros/anatomía & histología , Perros/clasificación , Extremidades/anatomía & histología , Estudio de Asociación del Genoma Completo , Glicoproteínas/genética , Glicoproteínas/fisiología , Proteína HMGA2/genética , Proteína HMGA2/fisiología , Cabello/anatomía & histología , Cardiopatías/genética , Cardiopatías/veterinaria , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/fisiología , Síndromes Neoplásicos Hereditarios/genética , Síndromes Neoplásicos Hereditarios/veterinaria , Osteosarcoma/genética , Osteosarcoma/veterinaria , Fenotipo , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo , Selección Genética , Piel/anatomía & histología , Cráneo/anatomía & histología , Proteína Smad2/genética , Proteína Smad2/fisiología , Especificidad de la Especie , Cola (estructura animal)/anatomía & histología
18.
Cancer Res ; 73(10): 3041-50, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23536553

RESUMEN

Embryonic rhabdomyosarcoma (ERMS) is the most common soft-tissue tumor in children. Here, we report the identification of the minor groove DNA-binding factor high mobility group AT-hook 2 (HMGA2) as a driver of ERMS development. HMGA2 was highly expressed in normal myoblasts and ERMS cells, where its expression was essential to maintain cell proliferation, survival in vitro, and tumor outgrowth in vivo. Mechanistic investigations revealed that upregulation of the insulin-like growth factor (IGF) mRNA-binding protein IGF2BP2 was critical for HMGA2 action. In particular, IGF2BP2 was essential for mRNA and protein stability of NRAS, a frequently mutated gene in ERMS. shRNA-mediated attenuation of NRAS or pharmacologic inhibition of the MAP-ERK kinase (MEK)/extracellular signal-regulated kinase (ERK) effector pathway showed that NRAS and NRAS-mediated signaling was required for tumor maintenance. Taken together, these findings implicate the HMGA2-IGFBP2-NRAS signaling pathway as a critical oncogenic driver in ERMS.


Asunto(s)
GTP Fosfohidrolasas/fisiología , Proteína HMGA2/fisiología , Proteínas de la Membrana/fisiología , Proteínas de Unión al ARN/fisiología , Rabdomiosarcoma Embrionario/etiología , Animales , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Humanos , Ratones , Mioblastos/química , Rabdomiosarcoma Embrionario/patología , Transducción de Señal
19.
Br J Haematol ; 156(3): 383-7, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22017592

RESUMEN

Patients with paroxysmal nocturnal haemoglobinuria (PNH) have expanded clonal cells bearing a somatic mutation in the PIGA gene. Our previous study on two PNH patients with chromosome 12 rearrangements demonstrated the involvement of HMGA2 expression in clonal expansion. The present study investigated HMGA2 expression in PNH patients without chromosomal abnormalities. The expression of short HMGA2 with latent exon was significantly high in peripheral blood cells from 18 of 24 patients. Over-expression of truncated HMGA2 in mouse bone marrow cells caused expansion in recipient mice. These results support the idea that deregulated expression of HMGA2 causes expansion of PNH cells.


Asunto(s)
Proteína HMGA2/fisiología , Hemoglobinuria Paroxística/metabolismo , Empalme Alternativo , Anemia Aplásica/patología , Animales , Células de la Médula Ósea/patología , División Celular , Células Cultivadas/trasplante , Células Clonales/patología , Ensayo de Unidades Formadoras de Colonias , Exones/genética , Regulación de la Expresión Génica , Proteína HMGA2/biosíntesis , Proteína HMGA2/genética , Hemoglobinuria Paroxística/genética , Hemoglobinuria Paroxística/patología , Humanos , Proteínas de la Membrana/deficiencia , Ratones , Quimera por Radiación , Proteínas Recombinantes de Fusión/fisiología , Transducción Genética
20.
Blood ; 117(22): 5860-9, 2011 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-21460244

RESUMEN

Overexpression of high mobility group AT-hook 2 (HMGA2) is found in a number of benign and malignant tumors, including the clonal PIGA(-) cells in 2 cases of paroxysmal nocturnal hemoglobinuria (PNH) and some myeloproliferative neoplasms (MPNs), and recently in hematopoietic cell clones resulting from gene therapy procedures. In nearly all these cases overexpression is because of deletions or translocations that remove the 3' untranslated region (UTR) which contains binding sites for the regulatory micro RNA let-7. We were therefore interested in the effect of HMGA2 overexpression in hematopoietic tissues in transgenic mice (ΔHmga2 mice) carrying a 3'UTR-truncated Hmga2 cDNA. ΔHmga2 mice expressed increased levels of HMGA2 protein in various tissues including hematopoietic cells and showed proliferative hematopoiesis with increased numbers in all lineages of peripheral blood cells, hypercellular bone marrow (BM), splenomegaly with extramedullary erythropoiesis and erythropoietin-independent erythroid colony formation. ΔHmga2-derived BM cells had a growth advantage over wild-type cells in competitive repopulation and serial transplantation experiments. Thus overexpression of HMGA2 leads to proliferative hematopoiesis with clonal expansion at the stem cell and progenitor levels and may account for the clonal expansion in PNH and MPNs and in gene therapy patients after vector insertion disrupts the HMGA2 locus.


Asunto(s)
Regiones no Traducidas 3'/genética , ADN Complementario/genética , Proteína HMGA2/fisiología , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Trastornos Mieloproliferativos/etiología , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Western Blotting , Médula Ósea/metabolismo , Médula Ósea/patología , Células Clonales , Eritropoyetina/sangre , Citometría de Flujo , Perfilación de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Humanos , Técnicas para Inmunoenzimas , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Trastornos Mieloproliferativos/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...