Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Cell Death Dis ; 15(2): 158, 2024 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-38383528

RESUMEN

Chemotherapy is a primary treatment for esophageal squamous cell carcinoma (ESCC). Resistance to chemotherapeutic drugs is an important hurdle to effective treatment. Understanding the mechanisms underlying chemotherapy resistance in ESCC is an unmet medical need to improve the survival of ESCC. Herein, we demonstrate that ferroptosis triggered by inhibiting high mobility group AT-hook 1 (HMGA1) may provide a novel opportunity to gain an effective therapeutic strategy against chemoresistance in ESCC. HMGA1 is upregulated in ESCC and works as a key driver for cisplatin (DDP) resistance in ESCC by repressing ferroptosis. Inhibition of HMGA1 enhances the sensitivity of ESCC to ferroptosis. With a transcriptome analysis and following-up assays, we demonstrated that HMGA1 upregulates the expression of solute carrier family 7 member 11 (SLC7A11), a key transporter maintaining intracellular glutathione homeostasis and inhibiting the accumulation of malondialdehyde (MDA), thereby suppressing cell ferroptosis. HMGA1 acts as a chromatin remodeling factor promoting the binding of activating transcription factor 4 (ATF4) to the promoter of SLC7A11, and hence enhancing the transcription of SLC7A11 and maintaining the redox balance. We characterized that the enhanced chemosensitivity of ESCC is primarily attributed to the increased susceptibility of ferroptosis resulting from the depletion of HMGA1. Moreover, we utilized syngeneic allograft tumor models and genetically engineered mice of HMGA1 to induce ESCC and validated that depletion of HMGA1 promotes ferroptosis and restores the sensitivity of ESCC to DDP, and hence enhances the therapeutic efficacy. Our finding uncovers a critical role of HMGA1 in the repression of ferroptosis and thus in the establishment of DDP resistance in ESCC, highlighting HMGA1-based rewiring strategies as potential approaches to overcome ESCC chemotherapy resistance. Schematic depicting that HMGA1 maintains intracellular redox homeostasis against ferroptosis by assisting ATF4 to activate SLC7A11 transcription, resulting in ESCC resistance to chemotherapy.


Asunto(s)
Neoplasias Esofágicas , Carcinoma de Células Escamosas de Esófago , Ferroptosis , Animales , Ratones , Carcinoma de Células Escamosas de Esófago/tratamiento farmacológico , Carcinoma de Células Escamosas de Esófago/genética , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , Proteína HMGA1a/genética , Resistencia a Antineoplásicos/genética , Ferroptosis/genética , Proteína HMGA1b , Línea Celular Tumoral
2.
Cell Death Dis ; 14(2): 140, 2023 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-36805591

RESUMEN

Analyses of several databases showed that the lncRNA RNF157 Antisense RNA 1 (RNF157-AS1) is overexpressed in epithelial ovarian cancer (EOC) tissues. In our study, suppressing RNF157-AS1 strikingly reduced the proliferation, invasion, and migration of EOC cells compared with control cells, while overexpressing RNF157-AS1 greatly increased these effects. By RNA pulldown assays, RNA binding protein immunoprecipitation (RIP) assays, and mass spectrometry, RNF157-AS1 was further found to be able to bind to the HMGA1 and EZH2 proteins. Chromatin immunoprecipitation (ChIP) assays showed that RNF157-AS1 and HMGA1 bound to the ULK1 promoter and prevented the expression of ULK1. Additionally, RNF157-AS1 interacted with EZH2 to bind to the DIRAS3 promoter and diminish DIRAS3 expression. ULK1 and DIRAS3 were found to be essential for autophagy. Combination autophagy inhibitor and RNF157-AS1 overexpression or knockdown, a change in the LC3 II/I ratio was found using immunofluorescence (IF) staining and western blot (WB) analysis. The autophagy level also was confirmed by autophagy/cytotoxicity dual staining. However, the majority of advanced EOC patients require platinum-based chemotherapy, since autophagy is a cellular catabolic response to cell stress. As a result, RNF157-AS1 increased EOC cell sensitivity to chemotherapy and death under cis-platinum (DDP) treatment by suppressing autophagy, as confirmed by cell count Kit-8 (CCK8) assays, flow cytometry, and autophagy/cytotoxicity dual staining. Therefore, the OS and PPS times were longer in EOC patients with elevated RNF157-AS1 expression. RNF157-AS1-mediated autophagy has potential clinical significance in DDP chemotherapy for EOC patients.


Asunto(s)
Neoplasias Ováricas , ARN Largo no Codificante , Femenino , Humanos , Carcinoma Epitelial de Ovario/genética , ARN Largo no Codificante/genética , Proteína HMGA1a , Autofagia/genética , Inmunoprecipitación de Cromatina , Proteína HMGA1b , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Proteínas de Unión al GTP rho , Homólogo de la Proteína 1 Relacionada con la Autofagia/genética , Péptidos y Proteínas de Señalización Intracelular/genética
3.
Cell Death Dis ; 12(5): 422, 2021 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-33927188

RESUMEN

Tumor-associated macrophages (TAMs) in the tumor microenvironment contribute to poor prognosis in gastric cancer (GC). However, the underlying mechanism by which TAMs promote GC progression and metastasis remains elusive. Expression of POU1F1 was detected in 60 matched GC-normal tissue pairs using qRT-PCR and immunohistochemistry (IHC) analysis. The correlation between POU1F1 and the clinical-pathological factors of GC patients were further assessed. Cell proliferation was monitored by CCK-8, colony formation, and 5-Ethynyl-2'-deoxyuridine (EdU) incorporation assays. Cell migration and invasion were assessed by transwell assays. The impact on angiogenesis was evaluated by tube formation assay. Xenograft model was generated to investigate the role of POU1F1 on tumor growth and lung metastasis in vivo. GST pull-down and Co-immunoprecipitation (Co-IP) were used to study the interaction between HMGA1B/2 and POU1F1. Chromatin immunoprecipitation (ChIP) and dual luciferase reporter assays were performed to investigate the transcriptional regulation of POU1F1. Flow cytometry was performed to detect the surface expression of macrophage markers. Upregulated POU1F1 observed both in GC tissues and cell lines was positively correlated with poor prognosis. Knockdown of POU1F1 inhibited cell proliferation, migration, invasion, and angiogenesis in vitro, and suppressed tumor growth in vivo. HMGA1B/2 transcriptionally activated-POU1F1. POU1F1 promoted GC progression via regulating macrophage proliferation, migration, polarization, and angiogenesis in a CXCL12/CXCR4-dependent manner. POU1F1 also promoted GC metastasis in lung by modulating macrophage polarization through CXCL12/CXCR4 axis in vivo. HMGA1B/2-upregulated POU1F1 promoted GC metastasis via regulating macrophage polarization in a CXCL12/CXCR4-dependent manner.


Asunto(s)
Quimiocina CXCL12/metabolismo , Proteína HMGA1b/metabolismo , Proteína HMGA2/metabolismo , Macrófagos/metabolismo , Receptores CXCR4/metabolismo , Neoplasias Gástricas/metabolismo , Factor de Transcripción Pit-1/metabolismo , Animales , Proliferación Celular/fisiología , Proteína HMGA1b/genética , Proteína HMGA2/genética , Xenoinjertos , Humanos , Activación de Macrófagos , Macrófagos/inmunología , Macrófagos/patología , Masculino , Ratones , Ratones Desnudos , Metástasis de la Neoplasia , Transducción de Señal , Neoplasias Gástricas/genética , Neoplasias Gástricas/inmunología , Neoplasias Gástricas/patología , Análisis de Supervivencia , Factor de Transcripción Pit-1/genética , Transfección , Microambiente Tumoral
4.
Nanomedicine ; 15(1): 231-242, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30308301

RESUMEN

Even if cancer specific biomarkers are present in peripheral blood of cancer patients, it is very difficult to detect them with conventional technology because of their low concentration. A potential cancer biomarker is the HMGA1b protein, whose overexpression is a feature of several human malignant neoplasias. By taking advantage of the surface plasmon resonance (SPR) phenomenon, we realized a specific nano/technology-based assay for cancer detection. More in details, anti-HMGA1b monoclonal antibodies, whose affinity was previously defined by ELISA, were immobilized onto metallic surfaces to develop a direct SPR-based assay. After having analyzed blood samples from colorectal cancer patients and healthy people for the presence of HMGA1b, we observed a 2-fold increase of the HMGA1b levels in the blood of cancer patients with respect to the healthy control people. We conclude that the set-up technology might allow to detect a tumoral mass through the evaluation of HMGA1b protein blood levels.


Asunto(s)
Biomarcadores de Tumor/sangre , Técnicas Biosensibles/métodos , Neoplasias Colorrectales/sangre , Proteína HMGA1b/sangre , Nanotecnología/métodos , Proteínas Recombinantes/inmunología , Biomarcadores de Tumor/inmunología , Estudios de Casos y Controles , Neoplasias Colorrectales/inmunología , Ensayo de Inmunoadsorción Enzimática , Proteína HMGA1b/inmunología , Humanos , Resonancia por Plasmón de Superficie
5.
Appl Immunohistochem Mol Morphol ; 25(4): 244-250, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-26657872

RESUMEN

BACKGROUND: High-mobility group proteins A (HMGA) are more abundant in rapidly dividing and transformed cells. These are a group of proteins regulating tumorigenesis and tumor invasion. Increased expression of HMGA1 and HMGA2 has been reported in various benign and malignant tumors. The aim of the present study was to analyze expression of HMGA1 and HMGA2 proteins in retinoblastoma. METHODS: Protein expression of HMGA1 and HMGA2 in 80 formalin-fixed retinoblastoma tissues was performed by immunohistochemistry, and their mRNA expressions were analyzed on 40 fresh primary enucleated retinoblastoma samples by semiquantitative reverse transcription polymerase chain reaction. Results were then correlated with clinicopathologic parameters. RESULTS: Immunohistochemical analysis of HMGA1 and HMGA2 was seen in 56.25% and 58.75% of retinoblastoma cases, respectively. mRNA expressions of HMGA1 and HMGA2 was found to be 57.55% and 62.5%, respectively. The mRNA results correlated well with immunostaining results. Expression of both HMGA1 and HMGA2 was significantly associated with choroidal invasion and poor tumor differentiation. CONCLUSIONS: HMGA1 and HMGA2 proteins may contribute to tumorigenesis of Rb. Expression of HMGA1 and HMGA2 predicts poor prognosis and could serve as a therapeutic target in the management of RB. Further experiments are needed to determine the role of these proteins as therapeutic targets in tumorigenesis.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Proteína HMGA1a/metabolismo , Proteína HMGA1b/metabolismo , Inmunohistoquímica/métodos , Isoformas de Proteínas/metabolismo , Neoplasias de la Retina/diagnóstico , Retinoblastoma/diagnóstico , Carcinogénesis , Diferenciación Celular , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino
6.
Oncotarget ; 7(19): 28724-35, 2016 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-26895108

RESUMEN

Pseudogenes are DNA sequences with high homology to the corresponding functional gene, but, because of the accumulation of various mutations, they have lost their initial functions to code for proteins. Consequently, pseudogenes have been considered until few years ago dysfunctional relatives of the corresponding ancestral genes, and then useless in the course of genome evolution. However, several studies have recently established that pseudogenes are owners of key biological functions. Indeed, some pseudogenes control the expression of functional genes by competitively binding to the miRNAs, some of them generate small interference RNAs to negatively modulate the expression of functional genes, and some of them even encode functional mutated proteins. Here, we concentrate our attention on the pseudogenes of the HMGA1 gene, that codes for the HMGA1a and HMGA1b proteins having a critical role in development and cancer progression. In this review, we analyze the family of HMGA1 pseudogenes through three aspects: classification, characterization, and their possible function and involvement in cancer.


Asunto(s)
Proteína HMGA1a/genética , Proteína HMGA1b/genética , Neoplasias/genética , Seudogenes/genética , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Proteína HMGA1a/metabolismo , Proteína HMGA1b/metabolismo , Humanos , Modelos Genéticos , Mutación , Neoplasias/metabolismo , Neoplasias/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo
7.
Acta Biochim Biophys Sin (Shanghai) ; 47(11): 880-9, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26423116

RESUMEN

A large number of calmodulin-like (CML) proteins are present in plants, but there is little detailed information on the functions of these proteins in rice (Oryza sativa L.). Here, the CML3 protein from rice (OsCML3) and its truncated form lacking the C-terminal extension (OsCML3m) were found to exhibit a Ca2+-binding property and subsequent conformational change, but the ability to bind the CaM kinase II peptide was only observed for OsCML3m. Changes in their secondary structure upon Ca2+-binding measured by circular dichroism revealed that OsCML3m had a higher helical content than OsCML3. Moreover, OsCML3 was mainly localized in the plasma membrane, whereas OsCML3m was found in the nucleus. The rice high mobility group B1 (OsHMGB1) protein was identified as one of the putative OsCML3 target proteins. Bimolecular fluorescence complementation analysis revealed that OsHMGB1 bound OsCML3, OsCML3m or OsCML3s (cysteine to serine mutation at the prenylation site) in the nucleus presumably through the methionine and phenylalanine-rich hydrophobic patches, confirming that OsHMGB1 is a target protein in planta. The effect of OsCML3 or OsCML3m on the DNA-binding ability of OsHMGB1 was measured using an electrophoretic mobility shift assay. OsCML3m decreased the level of OsHMGB1 binding to pUC19 double-stranded DNA whereas OsCML3 did not. Taken together, OsCML3 probably provides a mechanism for manipulating the DNA-binding ability of OsHMGB1 in the nucleus and its C-terminal extension provides an intracellular Ca2+ regulatory switch.


Asunto(s)
Señalización del Calcio , Calmodulina/metabolismo , Proteína HMGA1b/metabolismo , Oryza/metabolismo , Proteínas de Plantas/metabolismo , Calmodulina/química , Calmodulina/genética , Proteínas de Unión al ADN/metabolismo , Proteína HMGA1b/genética , Proteínas de Plantas/química , Proteínas de Plantas/genética
8.
Oncotarget ; 6(5): 2680-92, 2015 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-25595895

RESUMEN

Several recent studies have reported the Polycomb Repressive Complex 1 member CBX7 as a tumor-suppressor gene whose expression progressively decreases in different human carcinomas in relation with tumor grade, malignant stage and poor prognosis. We have previously demonstrated that CBX7 is able to inhibit the expression of the SPP1 gene, encoding the chemokine osteopontin that is over-expressed in cancer and has a critical role in cancer progression. Here, we have analyzed the mechanism by which CBX7 regulates the SPP1 gene expression. We show that the SPP1 transcriptional regulation mechanism involves the CBX7-interacting protein HMGA1b, that acts as a positive regulator of the SPP1 gene. In fact, we demonstrate that, in contrast with the transcriptional activity of CBX7, HMGA1b is able to increase the SPP1 expression by inducing the activity of its promoter. Moreover, we show that CBX7 interferes with HMGA1b on the SPP1 promoter and counteracts the positive transcriptional activity of HMGA1b on the SPP1 expression. Furthermore, since we found that also the NF-κB complex resulted involved in the modulation of the SPP1 expression in thyroid cells, we suppose that CBX7/HMGA1b/NF-κB could take part in the same transcriptional mechanism that finally leads to the regulation of the SPP1 gene expression. Taken together, our data show the important role played by CBX7 in the negative regulation of the SPP1 gene expression, thus contributing to prevent the acquisition of a malignant phenotype.


Asunto(s)
Carcinoma/metabolismo , Proteína HMGA1b/metabolismo , Osteopontina/metabolismo , Complejo Represivo Polycomb 1/metabolismo , Neoplasias de la Tiroides/metabolismo , Sitios de Unión , Carcinoma/genética , Carcinoma/patología , Carcinoma Papilar , Línea Celular Tumoral , Movimiento Celular , Regulación de la Expresión Génica , Células HEK293 , Proteína HMGA1b/genética , Humanos , FN-kappa B/genética , FN-kappa B/metabolismo , Osteopontina/genética , Complejo Represivo Polycomb 1/genética , Regiones Promotoras Genéticas , Transducción de Señal , Cáncer Papilar Tiroideo , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/patología , Transcripción Genética , Transfección
9.
BMC Cancer ; 14: 694, 2014 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-25245141

RESUMEN

BACKGROUND: Humans and dogs are affected by squamous cell carcinomas of the oral cavity (OSCC) in a considerably high frequency. The high mobility group A2 (HMGA2) protein was found to be highly expressed in human OSCC and its expression was suggested to act as a useful predictive and prognostic tool in clinical management of oral carcinomas. Herein the expression of HMGA2 and its sister gene HMGA1 were analysed within human and canine OSCC samples. Additionally, the HMGA negatively regulating miRNAs of the let-7 family as well as the let-7 regulating gene Lin28 were also comparatively analysed. Deregulations of either one of these members could affect the progression of human and canine OSCC. METHODS: Expression levels of HMGA1, HMGA2, Lin28, let-7a and mir-98 were analysed via relative qPCR in primary human and canine OSCC, thereof derived cell lines and non-neoplastic samples. Additionally, comparative HMGA2 protein expression was analysed by immunohistochemistry. RESULTS: In both species, a significant up-regulation of the HMGA2 gene was found within the neoplastic samples while HMGA1 expression did not show significant deregulations. Comparative analyses showed down-regulation of mir-98 in human samples and up-regulation of let-7a and mir-98 in canine neoplastic samples. HMGA2 immunostainings showed higher intensities within the invasive front of the tumours than in the centre of the tumour in both species. CONCLUSIONS: HMGA2 could potentially serve as tumour marker in both species while HMGA1 might play a minor role in OSCC progression. Comparative studies indicate an inverse correlation of HMGA2 and mir-98 expression in human samples whereas in dogs no such characteristic could be found.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma de Células Escamosas/veterinaria , Enfermedades de los Perros/metabolismo , Proteína HMGA2/metabolismo , MicroARNs/metabolismo , Neoplasias de la Boca/veterinaria , Proteínas de Unión al ARN/metabolismo , Adulto , Anciano , Animales , Biomarcadores de Tumor/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Línea Celular Tumoral , Enfermedades de los Perros/genética , Perros , Femenino , Expresión Génica , Proteína HMGA1a/genética , Proteína HMGA1a/metabolismo , Proteína HMGA1b/genética , Proteína HMGA1b/metabolismo , Proteína HMGA2/genética , Humanos , Masculino , MicroARNs/genética , Persona de Mediana Edad , Neoplasias de la Boca/genética , Neoplasias de la Boca/metabolismo , Proteínas de Unión al ARN/genética
10.
Expert Opin Ther Targets ; 18(5): 541-53, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24684280

RESUMEN

INTRODUCTION: Emerging evidence demonstrates that the high mobility group A1 (HMGA1) chromatin remodeling protein is a key molecular switch required by cancer cells for tumor progression and a poorly differentiated, stem-like state. Because the HMGA1 gene and proteins are expressed at high levels in all aggressive tumors studied to date, research is needed to determine how to 'turn off' this master regulatory switch in cancer. AREAS COVERED: In this review, we describe prior studies that underscore the central role of HMGA1 in refractory cancers and we discuss approaches to target HMGA1 in cancer therapy. EXPERT OPINION: Given the widespread overexpression of HMGA1 in diverse, aggressive tumors, further research to develop technology to target HMGA1 holds immense promise as potent anticancer therapy. Previous work in preclinical models indicates that delivery of short hairpin RNA or interfering RNA molecules to 'switch off' HMGA1 expression dramatically impairs cancer cell growth and tumor progression. The advent of nanoparticle technology to systemically deliver DNA or RNA molecules to tumors brings this approach even closer to clinical applications, although further efforts are needed to translate these advances into therapies for cancer patients.


Asunto(s)
Antineoplásicos/farmacología , Proteína HMGA1a/antagonistas & inhibidores , Proteína HMGA1b/antagonistas & inhibidores , Terapia Molecular Dirigida/métodos , Proteínas de Neoplasias/antagonistas & inhibidores , Animales , Antibióticos Antineoplásicos/farmacología , Antibióticos Antineoplásicos/uso terapéutico , Antineoplásicos/uso terapéutico , Diferenciación Celular , Cromatina/ultraestructura , Ensayos Clínicos como Asunto , Inhibidores de la Ciclooxigenasa 2/farmacología , Inhibidores de la Ciclooxigenasa 2/uso terapéutico , Progresión de la Enfermedad , Ensayos de Selección de Medicamentos Antitumorales , Flavonoides/farmacología , Flavonoides/uso terapéutico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Terapia Genética , Proteína HMGA1a/genética , Proteína HMGA1a/fisiología , Proteína HMGA1b/genética , Proteína HMGA1b/fisiología , Humanos , Ratones , Nanopartículas/administración & dosificación , Invasividad Neoplásica/fisiopatología , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Neoplasias/tratamiento farmacológico , Oxazinas/farmacología , Oxazinas/uso terapéutico , Piperidinas/farmacología , Piperidinas/uso terapéutico , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/farmacología , ARN Interferente Pequeño/uso terapéutico , Células Madre/citología , Células Madre/efectos de los fármacos
11.
Anticancer Res ; 33(10): 4261-6, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24122990

RESUMEN

BACKGROUND: Oral leukoplakia (LPL) is considered a potentially malignant disorder in the oral cavity and the gastric tract. High mobility group A (HMGA) proteins are important in the transformation of normal cells into cancer cells, but there is a lack of knowledge on their importance in oral cancer development. The aim of the current project was to investigate HMGA expression in LPLs with different levels of dysplasia. MATERIALS AND METHODS: Biopsies were histologically processed to visualize the expression of HMGA1 and HMGA2 using immunohistochemistry. RESULTS: An increase of HMGA1-positive cells correlating to the degree of dysplasia was registered in the epithelium and in the connective tissue. HMGA2 expression was seen in the epithelium and in the connective tissue but with no obvious correlation to the level of dysplasia. CONCLUSION: This is, to our knowledge, the first study showing the expression of HMGA proteins in healthy and non-healthy oral mucosa.


Asunto(s)
Proteína HMGA1a/metabolismo , Proteína HMGA1b/metabolismo , Proteína HMGA2/metabolismo , Leucoplasia Bucal/metabolismo , Estudios de Casos y Controles , Humanos , Inmunohistoquímica , Mucosa Bucal/metabolismo , Mucosa Bucal/patología
12.
Cell Biochem Funct ; 31(3): 228-36, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22961697

RESUMEN

High-mobility group A1 (HMGA1) encodes proteins that act as mediators in viral integration, modification of chromatin structure, neoplastic transformation and metastatic progression. Because HMGA1 is overexpressed in most cancers and has transcriptional relationships with several Wnt-responsive genes, we explored the involvement of HMGA1 in Wnt/ß-catenin/TCF-4 signalling. In adenomatous polyposis coli (APC(Min/+)) mice, we observed significant up-regulation of HMGA1 mRNA and protein in intestinal tumours when compared with normal intestinal mucosa. Conversely, restoration of Wnt signalling by the zinc induction of wild-type APC resulted in HMGA1 down-regulation in HT-29 cells. Because APC mutations are associated with mobilization of the ß-catenin/TCF-4 transcriptional complex and subsequent activation of downstream oncogenic targets, we analyzed the 5'-flanking sequence of HMGA1 for putative TCF-4 binding elements. We identified two regions that specifically bind the ß-catenin/TCF-4 complex in vitro and in vivo, identifying HMGA1 as an immediate target of the ß-catenin/TCF-4 signalling pathway in colon cancer. Collectively, these findings strongly implicate Wnt/ß-catenin/TCF-4 signalling in regulating HMGA1 to further expand the extensive regulatory network affected by Wnt/ß-catenin/TCF-4 signalling.


Asunto(s)
Proteína HMGA1a/metabolismo , Proteína HMGA1b/metabolismo , Proteína 2 Similar al Factor de Transcripción 7/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Regiones no Traducidas 5' , Proteína de la Poliposis Adenomatosa del Colon/genética , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Animales , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Regulación hacia Abajo/efectos de los fármacos , Células HCT116 , Proteína HMGA1a/genética , Proteína HMGA1b/genética , Células HT29 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Regiones Promotoras Genéticas , Unión Proteica , Transducción de Señal , Proteína 2 Similar al Factor de Transcripción 7/genética , Activación Transcripcional/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Zinc/farmacología , beta Catenina/genética
13.
Mol Biosyst ; 7(4): 1336-46, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21336378

RESUMEN

The high mobility group A1 (HMGA1) are non-histone chromosomal proteins consisting of HMGA1a and HMGA1b which act as architectural transcription factors. Elevated levels of HMGA1 are reported in a number of human cancers and suggested as tumor markers. Due to their role in neoplastic transformation and tumor progression, we considered HMGA1 as a potential target for downregulation at the transcriptional level. The present paper deals with the binding of a widely used chemotherapeutic agent, adriamycin (ADM), to hmg a1 gene promoter (-304 to -284), 21RY, and its effect on the expression of hmga1 at the mRNA and protein levels and further its cytotoxic efficacy in A431 cells. A strong complex (21RY-ADM) formation caused hypochromic and bathochromic changes in UV-absorption, considerable spectral changes in circular dichroism of adriamycin and DNA and significant quenching of fluorescence emission of ADM. Thermodynamics of 21RY-ADM interaction was studied by isothermal titration and differential scanning calorimetric techniques that revealed the binding to be exothermic and favored by both negative enthalpy and positive entropy changes. Further, even low concentrations (10.3 nM) of ADM showed cytotoxicity on human squamous carcinoma cells (A431) and caused downregulation (by ∼ 70%) of hmga1 at mRNA and protein levels. Present findings clearly support the inhibitory effect of ADM on hmg a1 which quite probably is the consequence of its binding to the targeted region of hmg a1. Therefore, it appears that hmg a1 is a novel potential chemotherapeutic target in treating carcinomas of epithelial origin like prostate, breast, thyroid etc.


Asunto(s)
Antineoplásicos/farmacología , Doxorrubicina/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteína HMGA1a , Regiones Promotoras Genéticas , Antineoplásicos/química , Antineoplásicos/toxicidad , Secuencia de Bases , Carcinoma de Células Escamosas , Línea Celular Tumoral , Doxorrubicina/química , Doxorrubicina/toxicidad , Orden Génico , Proteína HMGA1a/genética , Proteína HMGA1a/metabolismo , Proteína HMGA1b/genética , Proteína HMGA1b/metabolismo , Humanos , Termodinámica
14.
Eur J Cancer ; 47(3): 470-8, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21044834

RESUMEN

HMGA1 gene rearrangements have been frequently described in human lipomas. In vitro studies suggest that HMGA1 proteins have a negative role in the control of adipocyte cell growth, and that HMGA1 gene truncation acts in a dominant-negative fashion. Therefore, to define better the role of the HMGA1 alterations in the generation of human lipomas, we generated mice carrying an Hmga1b truncated (Hmga1b/T) gene. These mice develop a giant phenotype together with a drastic expansion of the retroperitoneal and subcutaneous white adipose tissue. We show that the activation of the E2F pathway likely accounts, at least in part, for this phenotype. Interestingly, the Hmga1b/T mice also develop B-cell lymphomas similar to that occurring in Hmga1-knockout mice, supporting a dominant-negative role of the Hmga1b/T mutant also in vivo.


Asunto(s)
Gigantismo/genética , Proteína HMGA1b/genética , Lipomatosis/genética , Linfoma de Células B/genética , Adipocitos/patología , Animales , Proliferación Celular , Factor de Transcripción E2F1/metabolismo , Femenino , Proteína HMGA1b/metabolismo , Linfoma de Células B/metabolismo , Linfoma de Células B/patología , Masculino , Ratones , Ratones Transgénicos , Mutación/genética , Fenotipo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
Mol Cell Endocrinol ; 326(1-2): 19-24, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20347930

RESUMEN

The high mobility group AT-hook (HMGA) proteins, a family of DNA architectural factors, are highly expressed during embryogenesis and play a crucial role in several different biological processes, as well as in tumorigenesis of a wide range of tissues, including pituitary. Indeed, HMGA2 has been found rearranged and amplified in human prolactinomas, and transgenic mice overexpressing either Hmga1 or Hmga2 develop pituitary adenomas secreting prolactin and growth hormone. Here, we overview HMGA proteins in human tumours, focusing on pituitary adenomas and the mechanisms by which the HMGA proteins are involved in their onset and development. Different HMGA-dependent potential drives of pituitary oncogenesis are discussed as future research directions in the field.


Asunto(s)
Adenoma/etiología , Proteína HMGA2/fisiología , Proteínas Oncogénicas/fisiología , Neoplasias Hipofisarias/etiología , Adenoma/genética , Animales , Proteína HMGA1a/metabolismo , Proteína HMGA1b/metabolismo , Proteína HMGA1c/metabolismo , Proteína HMGA2/genética , Proteína HMGA2/metabolismo , Humanos , Ratones , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Neoplasias Hipofisarias/genética
16.
Cancer Invest ; 28(4): 340-9, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-19916743

RESUMEN

It is well known that HMGA1 group of non-histone chromosomal proteins are up-regulated in several human cancers. We studied the HMGA1 expression in squamous cell carcinoma of skin in mice followed by the treatment with Cisplatin, which is often used in combination therapies of cancers. A short course of Cisplatin treatment led to apoptotic cell death and downregulation (by 40%) of HMGA1. However, extended treatment of Cisplatin caused necrotic cell death; concomitantly HMGA1 expression decreased by 90%. Present results indicate a strong association of HMGA1 with Cisplatin-linked tumor regression. Therefore, HMGA1 could be a potential target in designing therapeutic strategies against cancer.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carcinoma de Células Escamosas/tratamiento farmacológico , Cisplatino/farmacología , Proteína HMGA1a/fisiología , Proteína HMGA1b/fisiología , Neoplasias Cutáneas/tratamiento farmacológico , Animales , Carcinoma de Células Escamosas/química , Carcinoma de Células Escamosas/patología , Femenino , Proteína HMGA1a/análisis , Proteína HMGA1a/genética , Proteína HMGA1b/análisis , Proteína HMGA1b/genética , Ratones , Neoplasias Cutáneas/química , Neoplasias Cutáneas/patología
17.
Biol Cell ; 101(9): 511-24, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19250063

RESUMEN

BACKGROUND INFORMATION: miRNAs (microRNAs) are a class of non-coding RNAs that inhibit gene expression by binding to recognition elements, mainly in the 3' UTR (untranslated region) of mRNA. A single miRNA can target several hundred mRNAs, leading to a complex metabolic network. miR-16 (miRNA-16), located on chromosome 13q14, is involved in cell proliferation and apoptosis regulation; it may interfere with either oncogenic or tumour suppressor pathways, and is implicated in leukaemogenesis. These data prompted us to search for and validate novel targets of miR-16. RESULTS: In the present study, by using a combined bioinformatics and molecular approach, we identified two novel putative targets of miR-16, caprin-1 (cytoplasmic activation/proliferation-associated protein-1) and HMGA1 (high-mobility group A1), and we also studied cyclin E which had been previously recognized as an miR-16 target by bioinformatics database. Using luciferase activity assays, we demonstrated that miR-16 interacts with the 3' UTR of the three target mRNAs. We showed that miR-16, in MCF-7 and HeLa cell lines, down-regulates the expression of caprin-1, HMGA1a, HMGA1b and cyclin E at the protein level, and of cyclin E, HMGA1a and HMGA1b at the mRNA levels. CONCLUSIONS: Taken together, our data demonstrated that miR-16 can negatively regulate two new targets, HMGA1 and caprin-1, which are involved in cell proliferation. In addition, we also showed that the inhibition of cyclin E expression was due, at least in part, to a decrease in its mRNA stability.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proliferación Celular , Proteína HMGA1a/metabolismo , MicroARNs/metabolismo , Secuencia de Aminoácidos , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Proteína HMGA1a/química , Proteína HMGA1a/genética , Proteína HMGA1b/química , Proteína HMGA1b/genética , Proteína HMGA1b/metabolismo , Humanos , MicroARNs/química , MicroARNs/genética , Datos de Secuencia Molecular , Unión Proteica , Alineación de Secuencia
18.
J Med Chem ; 52(5): 1247-50, 2009 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-19215087

RESUMEN

A three-dimensional pharmacophore model was generated utilizing a set of known inhibitors of c-Myc-Max heterodimer formation. The model successfully identified a set of structurally diverse compounds with potential inhibitory activity against c-Myc. Nine compounds were tested in vitro, and four displayed affinities in the micromolar range and growth inhibitory activity against c-Myc-overexpressing cells. These studies demonstrate the applicability of pharmacophore modeling to the identification of novel and potentially more puissant inhibitors of the c-Myc oncoprotein.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/química , Modelos Moleculares , Proteínas Proto-Oncogénicas c-myc/química , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Benzofuranos/química , Benzofuranos/farmacología , Línea Celular , Proliferación Celular/efectos de los fármacos , Dicroismo Circular , Ensayo de Cambio de Movilidad Electroforética , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Técnicas de Inactivación de Genes , Células HL-60 , Proteína HMGA1b/biosíntesis , Proteína HMGA1b/genética , Humanos , Unión Proteica , Multimerización de Proteína , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Piridinas/química , Piridinas/farmacología , Pirrolidinas/química , Pirrolidinas/farmacología , Ratas , Tiazoles/química , Tiazoles/farmacología
19.
Oncogene ; 28(11): 1432-42, 2009 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-19169275

RESUMEN

The High Mobility Group proteins HMGA1 are nuclear architectural factors that play a critical role in a wide range of biological processes. Since recent studies have identified the microRNAs (miRNAs) as important regulators of gene expression, modulating critical cellular functions such as proliferation, apoptosis and differentiation, the aim of our work was to identify the miRNAs that are physiologically regulated by HMGA1 proteins. To this purpose, we have analysed the miRNA expression profile of mouse embryonic fibroblasts (MEFs) carrying two, one or no Hmga1 functional alleles using a microarray (miRNA microarray). By this approach, we found a miRNA expression profile that differentiates Hmga1-null MEFs from the wild-type ones. In particular, a significant decrease in miR-196a-2, miR-101b, miR-331 and miR-29a was detected in homozygous Hmga1-knockout MEFs in comparison with wild-type cells. Consistently, these miRNAs are downregulated in most of the analysed tissues of Hmga1-null mice in comparison with the wild-type mice. ChIP assay shows that HMGA1 is able to bind regions upstream of these miRNAs. Moreover, we identified the HMGA2 gene product as a putative target of miR-196a-2, suggesting that HMGA1 proteins are able to downregulate the expression of the other member of the HMGA family through the regulation of the miR-196a-2 expression. Finally, ATXN1 and STC1 gene products have been identified as targets of miR-101b. Therefore, it is reasonable to hypothesize that HMGA1 proteins are involved in several functions by regulating miRNA expression.


Asunto(s)
Regulación de la Expresión Génica , Proteína HMGA1a/fisiología , Proteína HMGA1b/fisiología , MicroARNs/genética , Animales , Ataxina-1 , Ataxinas , Perfilación de la Expresión Génica , Glicoproteínas/genética , Glicoproteínas/fisiología , Proteína HMGA1a/genética , Proteína HMGA1b/genética , Ratones , MicroARNs/fisiología , Células 3T3 NIH , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología
20.
Int J Oncol ; 32(2): 289-305, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18202751

RESUMEN

The 'high mobility group' HMGA protein family consists of four members: HMGA1a, HMGA1b and HMGA1c, which result from translation of alternative spliced forms of one gene and HMGA2, which is encoded for by another gene. HMGA proteins are characterized by three DNA-binding domains, called AT-hooks, and an acidic carboxy-terminal tail. HMGA proteins are architectural transcription factors that both positively and negatively regulate the transcription of a variety of genes. They do not display direct transcriptional activation capacity, but regulate gene expression by changing the DNA conformation by binding to AT-rich regions in the DNA and/or direct interaction with several transcription factors. In this way, they influence a diverse array of normal biological processes including cell growth, proliferation, differentiation and death. Both HMGA1 and HMGA2 are hardly detectable in normal adult tissue but are abundantly and ubiquitously expressed during embryonic development. In malignant epithelial tumors as well as in leukemia, however, expression of HMGA1 is again strongly elevated to embryonic levels thus leading to ectopic expression of (fetal) target genes. HMGA2 overexpression also has a causal role in inducing neoplasia. Besides overexpression of full length HMGA proteins in different tumors, the HMGA genes are often involved in chromosomal rearrangements. Such translocations are mostly detected in benign tumors of mesenchymal origin and are believed to be one of the most common chromosomal rearrangements in human neoplasia. To provide clarity in the abundance of articles on this topic, this review gives a general overview of the nuclear functions and regulation of the HMGA genes and corresponding proteins.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Proteínas HMGA/fisiología , Proteína HMGA1a/fisiología , Proteína HMGA1b/fisiología , Proteína HMGA2/fisiología , Neoplasias/genética , Neoplasias/metabolismo , Secuencia de Aminoácidos , Diferenciación Celular , Proliferación Celular , Cromosomas/ultraestructura , Proteínas HMGA/metabolismo , Humanos , Modelos Biológicos , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA