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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
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
2.
Nature ; 480(7376): 254-8, 2011 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-22037311

RESUMEN

A full description of the human proteome relies on the challenging task of detecting mature and changing forms of protein molecules in the body. Large-scale proteome analysis has routinely involved digesting intact proteins followed by inferred protein identification using mass spectrometry. This 'bottom-up' process affords a high number of identifications (not always unique to a single gene). However, complications arise from incomplete or ambiguous characterization of alternative splice forms, diverse modifications (for example, acetylation and methylation) and endogenous protein cleavages, especially when combinations of these create complex patterns of intact protein isoforms and species. 'Top-down' interrogation of whole proteins can overcome these problems for individual proteins, but has not been achieved on a proteome scale owing to the lack of intact protein fractionation methods that are well integrated with tandem mass spectrometry. Here we show, using a new four-dimensional separation system, identification of 1,043 gene products from human cells that are dispersed into more than 3,000 protein species created by post-translational modification (PTM), RNA splicing and proteolysis. The overall system produced greater than 20-fold increases in both separation power and proteome coverage, enabling the identification of proteins up to 105 kDa and those with up to 11 transmembrane helices. Many previously undetected isoforms of endogenous human proteins were mapped, including changes in multiply modified species in response to accelerated cellular ageing (senescence) induced by DNA damage. Integrated with the latest version of the Swiss-Prot database, the data provide precise correlations to individual genes and proof-of-concept for large-scale interrogation of whole protein molecules. The technology promises to improve the link between proteomics data and complex phenotypes in basic biology and disease research.


Asunto(s)
Isoformas de Proteínas/análisis , Isoformas de Proteínas/química , Proteoma/análisis , Proteoma/química , Proteómica/métodos , Empalme Alternativo , Línea Celular , Senescencia Celular/genética , Daño del ADN , Bases de Datos de Proteínas , Proteína HMGA1a/análisis , Proteína HMGA1b/análisis , Células HeLa , Humanos , Fenotipo , Procesamiento Proteico-Postraduccional , Proteolisis , Proteómica/instrumentación
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
Cell Death Differ ; 13(9): 1554-63, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16341121

RESUMEN

HMGA gene overexpression and rearrangements are frequent in several tumours, but their oncogenic function is still unclear. Here we report of a physical and functional interaction between High Mobility Group A1 (HMGA1) protein and p53 oncosuppressor. We found that HMGA1 binds p53 in vitro and in vivo, and both proteins are present in the same complexes bound to the Bax gene promoter. HMGA1 interferes with the p53-mediated transcription of p53 effectors Bax and p21(waf1) while cooperates with p53 in the transcriptional activation of the p53 inhibitor mdm2. This transcriptional modulation is associated with a reduced p53-dependent apoptosis in cells expressing exogenous HMGA1 and p53, or in cells expressing endogenously the proteins and in which p53 was activated by UV-irradiation. Furthermore, antisense inhibition of HMGA1b expression dramatically increases the UV-induced p53-mediated apoptosis. These data define a new physical and functional interaction between HMGA1 and p53 that modulates transcription of p53 target genes and inhibits apoptosis.


Asunto(s)
Apoptosis , Proteína HMGA1b/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Línea Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Humanos , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Transcripción Genética , Activación Transcripcional , Rayos Ultravioleta , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
13.
J Am Soc Mass Spectrom ; 18(9): 1569-78, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17627840

RESUMEN

High mobility group (HMG) A1 proteins are subject to a number of post-translational modifications, which may regulate their function in gene transcription and other cellular processes. We examined, by using mass spectrometry, the acetylation of HMGA1a and HMGA1b proteins induced by histone acetyltransferases p300 and PCAF in vitro and in PC-3 human prostate cancer cells in vivo. It turned out that five lysine residues in HMGA1a, i.e., Lys-14, Lys-64, Lys-66, Lys-70, and Lys-73, could be acetylated by both p300 and PCAF. We further quantified the level of acetylation by analyzing, with LC-MS/MS, the proteolytic peptides of the in vitro or in vivo acetylated HMGA1 proteins where the unmodified lysine residues were chemically derivatized with a perdeuterated acetyl group. Quantification results revealed that p300 and PCAF exhibited different site preferences for the acetylation; the preference of p300 acetylation followed the order of Lys-64 approximately Lys-70 > Lys-66 > Lys-14 approximately Lys73, whereas the selectivity of PCAF acetylation followed the sequence of Lys-70 approximately Lys-73 > Lys-64 approximately Lys-66 > Lys-14. HMGA1b was acetylated in a very similar fashion as HMGA1a. We also demonstrated that C-terminal phosphorylation of HMGA1 proteins did not affect the in vitro acetylation of the two proteins by either p300 or PCAF. Moreover, we examined the acetylation of lysine residues in HMGA1a and HMGA1b isolated from PC-3 human prostate cancer cells. Our results showed that all the above five lysine residues were also acetylated in vivo, with Lys-64, Lys-66 and Lys-70 in HMGA1a exhibiting higher levels of acetylation than Lys-14 and Lys-73.


Asunto(s)
Proteína HMGA1a/metabolismo , Proteína HMGA1b/metabolismo , Histona Acetiltransferasas/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias de la Próstata/química , Neoplasias de la Próstata/metabolismo , Proteínas/metabolismo , Acetilación , Línea Celular Tumoral , Cromatografía Líquida de Alta Presión/métodos , Proteína HMGA1a/química , Proteína HMGA1b/química , Histona Acetiltransferasas/química , Humanos , Masculino , Proteínas de Neoplasias/química , Proteínas/química , Espectrometría de Masa por Ionización de Electrospray/métodos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos
14.
Mol Cell Biol ; 23(7): 2225-38, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12640109

RESUMEN

A drastic reduction in BRCA1 gene expression is a characteristic feature of aggressive sporadic breast carcinoma. However, the mechanisms underlying BRCA1 downregulation in breast cancer are not well understood. Here we report that both in vitro and in vivo HMGA1b protein binds to and inhibits the activity of both human and mouse BRCA1 promoters. Consistently, murine embryonic stem (ES) cells with the Hmga1 gene deleted display higher Brca1 mRNA and protein levels than do wild-type ES cells. Stable transfection of MCF-7 cells with the HMGA1b cDNA results in a decrease of BRCA1 gene expression and in a lack of BRCA1 induction after estrogen treatment. Finally, we found an inverse correlation between HMGA1 and BRCA1 mRNA and protein expression in human mammary carcinoma cell lines and tissues. These data indicate that HMGA1 proteins are involved in transcriptional regulation of the BRCA1 gene, and their overexpression may have a role in BRCA1 downregulation observed in aggressive mammary carcinomas.


Asunto(s)
Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Neoplasias de la Mama/metabolismo , Carcinoma/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Proteína HMGA1b/metabolismo , Regiones Promotoras Genéticas/fisiología , Animales , Proteína BRCA1/deficiencia , Neoplasias de la Mama/genética , Carcinoma/genética , Línea Celular , Estrógenos/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Marcación de Gen , Genes Reporteros , Proteína HMGA1b/genética , Proteína HMGA1b/farmacología , Humanos , Ratones , Regiones Promotoras Genéticas/efectos de los fármacos , Unión Proteica/genética , ARN Mensajero/metabolismo , Fase S , Células Madre/citología , Células Madre/metabolismo , Transfección , Células Tumorales Cultivadas
15.
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
16.
Nucleic Acids Res ; 31(17): 5025-32, 2003 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-12930952

RESUMEN

The high mobility group A (HMGA) family of non-histone chromosomal proteins is encoded by two related genes, HMGA1 and HMGA2. HMGA proteins are architectural transcription factors that have been found to regulate the transcription of a large number of genes. They are also some of the most commonly dysregulated genes in human neoplasias, highlighting a role in growth control. HMGA1 and HMGA2 have also been found to stimulate retroviral integration in vitro. In this study, we have cloned chicken HMGA1, and used the chicken DT40 B-cell lymphoma line to generate cells lacking HMGA1, HMGA2 and both in combination. We tested these lines for effects on cellular growth, gene control and retroviral integration. Surprisingly, we found that the HMGA gene family is dispensable for growth in DT40 cells, and that there is no apparent defect in retroviral integration in the absence of HMGA1 or HMGA2. We also analyzed the activity of approximately 4000 chicken genes, but found no significant changes. We conclude that HMGA proteins are not strictly required for growth control or retroviral integration in DT40 cells and may well be redundant with other factors.


Asunto(s)
Proteínas HMGA/genética , Familia de Multigenes/genética , Secuencia de Aminoácidos , Animales , División Celular/genética , Línea Celular , Línea Celular Tumoral , Embrión de Pollo , Pollos , Clonación Molecular , ADN Complementario/química , ADN Complementario/genética , Expresión Génica , Perfilación de la Expresión Génica , Vectores Genéticos/genética , Genotipo , Proteína HMGA1b/genética , Proteína HMGA2/genética , Humanos , Lentivirus/genética , Luciferasas/genética , Luciferasas/metabolismo , Datos de Secuencia Molecular , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Retroviridae/genética , Alineación de Secuencia , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido
17.
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
18.
Oncogene ; 23(25): 4466-76, 2004 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-15064752

RESUMEN

Activation of the transcription factor AP-1 (activator protein-1) is required for tumor promotion and maintenance of malignant phenotype. A number of AP-1-regulated genes that play a role in tumor progression have been identified. However, AP-1-regulated genes driving tumor induction are yet to be defined. Previous studies have established that expression of a dominant-negative c-Jun (TAM67) inhibits phorbol 12-tetradecanoyl-13-acetate (TPA)-induced AP-1 transactivation as well as transformation in mouse epidermal JB6/P+ cells and tumor promotion in mouse skin carcinogenesis. In this study, we utilized the tumor promotion-sensitive JB6/P+ cells to identify AP-1-regulated TAM67 target genes and to establish causal significance in transformation for one target gene. A 2700 cDNA microarray was queried with RNA from TPA-treated P+ cells with or without TAM67 expression. Under conditions in which TAM expression inhibited TPA-induced transformation, microarray analysis identified a subset of six genes induced by TPA and suppressed by TAM67. One of the identified genes, the high-mobility group protein A1 (Hmga1) is induced by TPA in P+, but not in transformation-resistant P cells. We show that TPA induction of the architectural transcription factor HMGA1 is inhibited by TAM67, is extracellular-signal-regulated kinase (ERK)-activation dependent, and is mediated by AP-1. HMGA1 antisense construct transfected into P+ cells blocked HMGA1 protein expression and inhibited TPA-induced transformation indicating that HMGA1 is required for transformation. HMGA1 is not however sufficient as HMGA1a or HMGA1b overexpression did not confer transformation sensitivity on P- cells. Although HMGA1 expression is ERK dependent, it is not the only ERK-dependent event required for transformation because it does not suffice to rescue ERK-deficient P- cells. Our study shows (a) TAM 67 when it inhibits AP-1 and transformation, targets a relatively small number of genes; (b) HMGA1, a TAM67 target gene, is causally related to transformation and therefore a potentially important target for cancer prevention.


Asunto(s)
Transformación Celular Neoplásica/genética , Células Epidérmicas , Genes jun/genética , Proteína HMGA1a/fisiología , Proteína HMGA1b/fisiología , Quinasa 1 de Quinasa de Quinasa MAP , Proteínas Proto-Oncogénicas c-jun/fisiología , Transcripción Genética/genética , Animales , Butadienos/farmacología , Línea Celular/efectos de los fármacos , Línea Celular/metabolismo , Transformación Celular Neoplásica/efectos de los fármacos , Células Clonales/efectos de los fármacos , Células Clonales/metabolismo , Ciclina D1/biosíntesis , Ciclina D1/genética , ADN Complementario/genética , Susceptibilidad a Enfermedades , Epidermis/efectos de los fármacos , Epidermis/metabolismo , Perfilación de la Expresión Génica , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Transgénicos , Proteína Quinasa 1 Activada por Mitógenos/fisiología , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/fisiología , Nitrilos/farmacología , Análisis de Secuencia por Matrices de Oligonucleótidos , Oligonucleótidos Antisentido/farmacología , Osteopontina , Proteínas Proto-Oncogénicas c-jun/deficiencia , Sialoglicoproteínas/biosíntesis , Sialoglicoproteínas/genética , Acetato de Tetradecanoilforbol/toxicidad , Factor de Transcripción AP-1/fisiología , Transcripción Genética/efectos de los fármacos
19.
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
20.
Gene ; 330: 93-9, 2004 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-15087128

RESUMEN

Due to the emerging advantages of numerous canine diseases as a genetic model for their human orthologs, the dog could join the mouse as the species of choice to unravel genetic mechanisms, e.g. of cancer predisposition, development and progression. However, precondition for such studies is the characterisation of the corresponding canine genes. Human and murine HMGA1 non-histone proteins participate in a wide variety of cellular processes including regulation of inducible gene transcription, integration of retroviruses into chromosomes, and the induction of neoplastic transformation and promotion of metastatic progression of cancer cells. Chromosomal aberrations affecting the human HMGA1 gene at 6p21 were described in several tumours like pulmonary chondroid hamartomas, uterine leiomyomas, follicular thyroid adenomas and others. Over-expression of the proteins of HMGA1 is characteristic for various malignant tumours suggesting a relation between high titer of the protein and the neoplastic phenotype. In this study, we characterised the molecular structure of the canine HMGA1 cDNA, its splice variants and predicted proteins HMGA1a and HMGA1b. Furthermore, we compared the coding sequence(s) (CDS) of both splice variants for 12 different breeds, screened them for single nucleotide polymorphisms (SNPs) and characterised a basic expression pattern.


Asunto(s)
Perros/genética , Proteína HMGA1a/genética , Proteína HMGA1b/genética , Empalme Alternativo , Secuencia de Aminoácidos , Animales , ADN Complementario/química , ADN Complementario/genética , Exones , Expresión Génica , Genes/genética , Intrones , Masculino , Datos de Secuencia Molecular , Alineación de Secuencia , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA