Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 115
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Front Cell Infect Microbiol ; 13: 1267288, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37842005

RESUMEN

Aim: This study established a high-throughput multiplex genetic detection assay (HMGA) for rapid identification, semi-quantification and virulence analysis of Helicobacter pylori directly from the clinical non-invasive oral samples. Methods: The gastric mucosa and oral samples were collected from 242 patients in Shanghai from 2021 to 2022. All the samples were detected by routine clinical tests for H. pylori and Sanger sequenced for inconsistent results. A new multiplex PCR assay providing results within 4 hours was designed and optimized involving fluorescent dye-labeled specific primers targeted 16S rRNA gene, semi-quantitative gene ureC and 10 virulence genes of H. pylori. Semi-quantification was carried out by simulating the serial 10-fold dilutions of positive oral samples, and the H. pylori loads in different clinical samples were further compared. The mixed plasmids of virulence genes vacA s1, vacA m1 and vacA m2 were used to evaluate the performance on different genotypes. The consistency of 10 virulence genes in gastric mucosa, saliva, mouthwash and dental plaque of H. pylori-positive patients was compared. Results: The non-invasive HMGA was highly specific for detection of all 12 targets of H. pylori and human internal reference gene ß-globin, and the sensitivity to all target genes could reach 10 copies/µL. Compared with routine clinical tests and sequencing, non-invasive HMGA has a high level (>0.98) of sensitivity, specificity, accuracy, PPV, NPV and kappa coefficient for direct detection of H. pylori in oral samples. Moreover, by detecting peak area levels of ureC, it was confirmed that the H. pylori loads in gastric mucosa were significantly higher than those of the three kinds of oral samples (p<0.05). We also found that 45.0% (91/202) of patients had different H. pylori virulence genes in different oral samples. The concordance of positive detection rates of each virulence gene between saliva and gastric mucosa was more than 78% (p<0.05). Conclusion: The non-invasive HMGA proved to be a reliable method for the rapid H. pylori identification, semi-quantification and detection of 10 virulence genes directly in oral samples, providing a new idea for non-invasive detection of H. pylori.


Asunto(s)
Proteínas HMGA , Infecciones por Helicobacter , Helicobacter pylori , Humanos , Proteínas Bacterianas/genética , Virulencia/genética , Genotipo , ARN Ribosómico 16S/genética , China , Proteínas HMGA/genética , Infecciones por Helicobacter/diagnóstico , Antígenos Bacterianos/genética
2.
Cell Cycle ; 21(12): 1335-1346, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35282770

RESUMEN

Neuroendocrine tumors (NETs) are neoplasms derived from neuroendocrine cells. One of their main features is to often remain asymptomatic and clinically undetectable. High Mobility Group A (HMGA) proteins belong to a family of non-histone chromatinic proteins able to modulate gene expression through the interaction with DNA and transcription factors. They are overexpressed in most of the human malignancies, playing a critical role in carcinogenesis. However, their expression levels and their role in neuroendocrine carcinogenesis has not been exhaustively evaluated until now. Therefore, in this study, we have addressed the validity of using the expression of HMGA1 as a diagnostic marker and have investigated its role in NET carcinogenesis. The expression of HMGA1 has been evaluated by qRT-PCR and immunohistochemistry, using NET tissue microarrays, in a cohort of gastroenteropancreatic (GEP)-NET samples. The expression levels of HMGA1 have been then correlated with the main clinical features of NET samples. Finally, the contribution of HMGA1 overexpression to NET development has been addressed as far as the modulation of proliferation and migration abilities of NET cells is concerned. Here, we report that HMGA1 is overexpressed in GEP-NET samples, at both mRNA and protein levels, and that the silencing of HMGA1 protein expression interferes with the ability of NET cells to proliferate and migrate through the downregulation of Cyclin E, Cyclin B1 and EZH2. These results propose the HMGA proteins as new diagnostic and prognostic markers.


Asunto(s)
Proteínas HMGA , Proteína HMGA1a/metabolismo , Tumores Neuroendocrinos , Carcinogénesis , Proteínas HMGA/genética , Proteína HMGA1a/genética , Humanos , Neoplasias Intestinales , Tumores Neuroendocrinos/genética , Tumores Neuroendocrinos/metabolismo , Tumores Neuroendocrinos/patología , Neoplasias Pancreáticas , Neoplasias Gástricas , Factores de Transcripción
3.
Cell Mol Life Sci ; 78(3): 817-831, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32920697

RESUMEN

The high mobility group A (HMGA) proteins are found to be aberrantly expressed in several tumors. Studies (in vitro and in vivo) have shown that HMGA protein overexpression has a causative role in carcinogenesis process. HMGA proteins regulate cell cycle progression through distinct mechanisms which strongly influence its normal dynamics along malignant transformation. Tumor protein p53 (TP53) is the most frequently altered gene in cancer. The loss of its activity is recognized as the fall of a barrier that enables neoplastic transformation. Among the different functions, TP53 signaling pathway is tightly involved in control of cell cycle, with cell cycle arrest being the main biological outcome observed upon p53 activation, which prevents accumulation of damaged DNA, as well as genomic instability. Therefore, the interaction and opposing effects of HMGA and p53 proteins on regulation of cell cycle in normal and tumor cells are discussed in this review. HMGA proteins and p53 may reciprocally regulate the expression and/or activity of each other, leading to the counteraction of their regulation mechanisms at different stages of the cell cycle. The existence of a functional crosstalk between these proteins in the control of cell cycle could open the possibility of targeting HMGA and p53 in combination with other therapeutic strategies, particularly those that target cell cycle regulation, to improve the management and prognosis of cancer patients.


Asunto(s)
Puntos de Control del Ciclo Celular/fisiología , Proteínas HMGA/metabolismo , Neoplasias/patología , Proteína p53 Supresora de Tumor/metabolismo , Daño del ADN , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Inestabilidad Genómica , Proteínas HMGA/genética , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/genética
4.
Molecules ; 25(24)2020 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-33327391

RESUMEN

A stable intense resistance called "nonhost resistance" generates a complete multiple-gene resistance against plant pathogenic species that are not pathogens of pea such as the bean pathogen, Fusarium solani f. sp. phaseoli (Fsph). Chitosan is a natural nonhost resistance response gene activator of defense responses in peas. Chitosan may share with cancer-treatment compounds, netropsin and some anti-cancer drugs, a DNA minor groove target in plant host tissue. The chitosan heptamer and netropsin have the appropriate size and charge to reside in the DNA minor groove. The localization of a percentage of administered radio-labeled chitosan in the nucleus of plant tissue in vivo indicates its potential to transport to site(s) within the nuclear chromatin (1,2). Other minor groove-localizing compounds administered to pea tissue activate the same secondary plant pathway that terminates in the production of the anti-fungal isoflavonoid, pisatin an indicator of the generated resistance response. Some DNA minor groove compounds also induce defense genes designated as "pathogenesis-related" (PR) genes. Hypothetically, DNA targeting components alter host DNA in a manner enabling the transcription of defense genes previously silenced or minimally expressed. Defense-response-elicitors can directly (a) target host DNA at the site of transcription or (b) act by a series of cascading events beginning at the cell membrane and indirectly influence transcription. A single defense response, pisatin induction, induced by chitosan and compounds with known DNA minor groove attachment potential was followed herein. A hypothesis is formulated suggesting that this DNA target may be accountable for a portion of the defense response generated in nonhost resistance.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Quitosano/farmacología , Sustancias Intercalantes/farmacología , Netropsina/farmacología , Pisum sativum/genética , Enfermedades de las Plantas/genética , Pterocarpanos/farmacología , Antineoplásicos Fitogénicos/química , Bencimidazoles/química , Bencimidazoles/farmacología , Quitosano/química , Cromatina/química , Cromatina/efectos de los fármacos , Cromatina/metabolismo , Cromomicinas/química , Cromomicinas/farmacología , ADN de Plantas/genética , ADN de Plantas/metabolismo , Resistencia a la Enfermedad/genética , Fusarium/crecimiento & desarrollo , Fusarium/patogenicidad , Regulación de la Expresión Génica de las Plantas , Proteínas HMGA/genética , Proteínas HMGA/metabolismo , Sustancias Intercalantes/química , Netropsina/química , Pisum sativum/inmunología , Pisum sativum/metabolismo , Pisum sativum/microbiología , Enfermedades de las Plantas/inmunología , Enfermedades de las Plantas/microbiología , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Pterocarpanos/química , Transcripción Genética
5.
Elife ; 92020 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-32965216

RESUMEN

During gastrulation, neural crest cells are specified at the neural plate border, as characterized by Pax7 expression. Using single-cell RNA sequencing coupled with high-resolution in situ hybridization to identify novel transcriptional regulators, we show that chromatin remodeler Hmga1 is highly expressed prior to specification and maintained in migrating chick neural crest cells. Temporally controlled CRISPR-Cas9-mediated knockouts uncovered two distinct functions of Hmga1 in neural crest development. At the neural plate border, Hmga1 regulates Pax7-dependent neural crest lineage specification. At premigratory stages, a second role manifests where Hmga1 loss reduces cranial crest emigration from the dorsal neural tube independent of Pax7. Interestingly, this is rescued by stabilized ß-catenin, thus implicating Hmga1 as a canonical Wnt activator. Together, our results show that Hmga1 functions in a bimodal manner during neural crest development to regulate specification at the neural plate border, and subsequent emigration from the neural tube via canonical Wnt signaling.


The neural plate is a structure that serves as the basis for the brain and central nervous system during the development of animals with a backbone. In particular, the tissues at the border of the neural plate become the neural crest, a group of highly mobile cells that can specialize to form nerves and parts of the face. The exact molecular mechanisms that allow the crest to emerge are still unknown. The protein Hmga1 alters how genes are packaged and organized inside cells, which in turn influences how genes are switched on and off. Here, Gandhi et al. studied how Hmga1 helps to shape the neural crest in developing chicken embryos. To do so, they harnessed a genetic tool called CRISPR-Cas9, and deleted the gene that encodes Hmga1 at specific developmental stages. This manipulation highlighted two periods where Hmga1 is active. First, Hmga1 helped to define neural crest cells at the neural plate border by activating a gene called pax7. Then, at a later stage, Hmga1 allowed these cells to move to other parts of the body by triggering the Wnt communication system. Failure for the neural crest to develop properly causes birth defects and cancers such as melanoma and childhood neuroblastoma, highlighting the need to better understand how this structure is formed. In addition, a better grasp of the roles of Hmga1 in healthy development could help to appreciate how it participates in a range of adult cancers.


Asunto(s)
Proteínas Aviares/genética , Movimiento Celular , Embrión de Pollo/embriología , Ensamble y Desensamble de Cromatina/fisiología , Proteínas HMGA/genética , Cresta Neural/embriología , Animales , Proteínas Aviares/metabolismo , Pollos/fisiología , Proteínas HMGA/metabolismo , Vía de Señalización Wnt
6.
Int J Mol Sci ; 21(2)2020 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-31963852

RESUMEN

HMGA (high mobility group A) (HMGA1 and HMGA2) are small non-histone proteins that can bind DNA and modify chromatin state, thus modulating the accessibility of regulatory factors to the DNA and contributing to the overall panorama of gene expression tuning. In general, they are abundantly expressed during embryogenesis, but are downregulated in the adult differentiated tissues. In the present review, we summarize some aspects of their role during development, also dealing with relevant studies that have shed light on their functioning in cell biology and with emerging possible involvement of HMGA1 and HMGA2 in evolutionary biology.


Asunto(s)
Proteínas HMGA/genética , Proteínas HMGA/metabolismo , Animales , Ciclo Celular , Ensamble y Desensamble de Cromatina , Desarrollo Embrionario , Evolución Molecular , Regulación del Desarrollo de la Expresión Génica , Humanos
7.
Int J Mol Sci ; 21(1)2020 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-31935816

RESUMEN

HMGA1 and HMGA2 are chromatin architectural proteins that do not have transcriptional activity per se, but are able to modify chromatin structure by interacting with the transcriptional machinery and thus negatively or positively regulate the transcription of several genes. They have been extensively studied in cancer where they are often found to be overexpressed but their functions under physiologic conditions have still not been completely addressed. Hmga1 and Hmga2 are expressed during the early stages of mouse development, whereas they are not detectable in most adult tissues. Hmga overexpression or knockout studies in mouse have pointed to a key function in the development of the embryo and of various tissues. HMGA proteins are expressed in embryonic stem cells and in some adult stem cells and numerous experimental data have indicated that they play a fundamental role in the maintenance of stemness and in the regulation of differentiation. In this review, we discuss available experimental data on HMGA1 and HMGA2 functions in governing embryonic and adult stem cell fate. Moreover, based on the available evidence, we will aim to outline how HMGA expression is regulated in different contexts and how these two proteins contribute to the regulation of gene expression and chromatin architecture in stem cells.


Asunto(s)
Células Madre Adultas/metabolismo , Diferenciación Celular , Células Madre Embrionarias/metabolismo , Proteínas HMGA/genética , Células Madre Adultas/citología , Animales , Ensamble y Desensamble de Cromatina , Células Madre Embrionarias/citología , Regulación del Desarrollo de la Expresión Génica , Proteínas HMGA/metabolismo , Humanos
8.
Int J Mol Sci ; 21(3)2020 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-31979076

RESUMEN

High mobility group A (HMGA) proteins are oncofoetal chromatin architectural factors that are widely involved in regulating gene expression. These proteins are unique, because they are highly expressed in embryonic and cancer cells, where they play a relevant role in cell proliferation, stemness, and the acquisition of aggressive tumour traits, i.e., motility, invasiveness, and metastatic properties. The HMGA protein expression levels and activities are controlled by a connected set of events at the transcriptional, post-transcriptional, and post-translational levels. In fact, microRNA (miRNA)-mediated RNA stability is the most-studied mechanism of HMGA protein expression modulation. In this review, we contribute to a comprehensive overview of HMGA-targeting miRNAs; we provide detailed information regarding HMGA gene structural organization and a comprehensive evaluation and description of HMGA-targeting miRNAs, while focusing on those that are widely involved in HMGA regulation; and, we aim to offer insights into HMGA-miRNA mutual cross-talk from a functional and cancer-related perspective, highlighting possible clinical implications.


Asunto(s)
Cromatina/genética , Proteínas HMGA/genética , MicroARNs/genética , Animales , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Neoplasias/genética
9.
Talanta ; 206: 120220, 2020 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-31514891

RESUMEN

This work addresses a technological advance applied to the construction of a magnetogenoassay with electrochemical transduction for the maize taxon-specific (HMGA gene) detection using gold-coated magnetic nanoparticles as nanosized platform. Superparamagnetic core-shell Fe3O4@Au nanoparticles (10.4 ±â€¯1.7 nm) were used to assemble the genoassay through the covalent immobilization of HMGA DNA probes onto carboxylated self-assembled monolayers at the nanoparticles surface. A hybridization reaction using sandwich format was selected to prevent inefficient hybridization connected with stable secondary DNA structures using also fluorescein isothiocyanate as DNA signaling tag. The labelling of the hybridization reaction with enzymes allowed the chronoamperometric measurement of the peroxidase activity linked to the nanoplatform located on gold surface. Using this electrochemical magnetogenoassay a linear concentration range from 0.5 to 5 nM and a LOD of 90 pM with a RSD <1.2% was calculated. Certified maize was evaluated without further purification after PCR amplification. This work highlights the efficacy of the electrochemical magnetogenoassay for the HMGA detection, showing its potential as alternative procedure for the verification of the compliance of the legislation.


Asunto(s)
Técnicas Biosensibles/métodos , Genes de Plantas , Oro/química , Proteínas HMGA/genética , Nanopartículas de Magnetita/química , Zea mays/genética , Secuencia de Bases , Sondas de ADN/química , Sondas de ADN/genética , ADN de Plantas/química , ADN de Plantas/genética , ADN de Cadena Simple/química , ADN de Cadena Simple/genética , Técnicas Electroquímicas/métodos , Ácidos Nucleicos Inmovilizados/química , Ácidos Nucleicos Inmovilizados/genética , Límite de Detección , Hibridación de Ácido Nucleico , Proteínas de Plantas/genética , Plantas Modificadas Genéticamente/genética
10.
Recent Pat Anticancer Drug Discov ; 14(3): 258-267, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31538905

RESUMEN

BACKGROUND: The high mobility group A proteins modulate the transcription of numerous genes by interacting with transcription factors and/or altering the structure of chromatin. These proteins are involved in both benign and malignant neoplasias as a result of several pathways. A large amount of benign human mesenchymal tumors has rearrangements of HMGA genes. On the contrary, malignant tumors show unarranged HMGA overexpression that is frequently and causally related to neoplastic cell transformation. Here, we review the function of the HMGA proteins in human neoplastic disorders, the pathways by which they contribute to carcinogenesis and the new patents focused on targeting HMGA proteins. OBJECTIVE: Current review was conducted to check the involvement of HMGA as a druggable target in cancer treatment. METHODS: We reviewed the most recent patents focused on targeting HMGA in cancer treatment analyzing patent literature published during the last years, including the World Intellectual Property Organization (WIPO®), United States Patent Trademark Office (USPTO®), Espacenet®, and Google Patents. RESULTS: HMGA proteins are intriguing targets for cancer therapy and are objects of different patents based on the use of DNA aptamers, inhibitors, oncolytic viruses, antisense molecules able to block their oncogenic functions. CONCLUSION: Powerful strategies able to selectively interfere with HMGA expression and function could represent a helpful approach in the development of new anti-cancer therapies.


Asunto(s)
Aptámeros de Nucleótidos/farmacología , Proteínas HMGA/antagonistas & inhibidores , Neoplasias/terapia , ARN Interferente Pequeño/farmacología , Bases de Datos Factuales , Proteínas HMGA/genética , Proteínas HMGA/metabolismo , Humanos , Terapia Molecular Dirigida , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Viroterapia Oncolítica , Patentes como Asunto , Factores de Transcripción/metabolismo
11.
Cancer Invest ; 37(8): 339-354, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31412717

RESUMEN

Squamous cell carcinoma (SCC) of skin has no standard treatment regimen, resulting in recurrences/metastasis. Although, doxorubicin (Dox), an anthracycline antibiotic has demonstrated some degree of efficacy. Molecular imaging can help in assessment of treatment response and prognosis of SCCs. MRI data showed that spin-spin relaxation (T2) time was longer (138 ± 2 msec) in Dox treated Test-II and there is no significant difference in spin-lattice relaxation (T1) time with respective controls. These findings further corroborated with the histology, proliferation index, apoptotic index, and HMGA1 protein expression. Thus, MRI may be a useful tool for monitoring treatment response noninvasively for skin tumor prognosis.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Carcinoma de Células Escamosas/tratamiento farmacológico , Doxorrubicina/farmacología , Imagen por Resonancia Magnética , Imagen Molecular/métodos , Neoplasias Cutáneas/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Biomarcadores/metabolismo , Carcinoma de Células Escamosas/diagnóstico por imagen , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Proteínas HMGA/genética , Proteínas HMGA/metabolismo , Ratones , Valor Predictivo de las Pruebas , Neoplasias Cutáneas/diagnóstico por imagen , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Factores de Tiempo , Carga Tumoral/efectos de los fármacos
12.
FASEB J ; 33(11): 12348-12363, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31415216

RESUMEN

Abnormal placental development is one of the main etiological factors for intrauterine growth restriction (IUGR). Here, we show that LIN28A and LIN28B are significantly lower and lethal-7 (let-7) microRNAs (miRNAs) significantly higher in term human IUGR vs. normal placentas. We hypothesize that let-7 miRNAs regulate genes with known importance for human placental development [high-mobility group AT-hook 1 (HMGA1), transcriptional regulator Myc-like (c-myc), vascular endothelial growth factor A (VEGF-A), and Wnt family member 1 (WNT1)] by targeting the AT-rich interacting domain (ARID)-3B complex. ACH-3P cells with LIN28A and LIN28B knockout (DKOs) significantly increased let-7 miRNAs, leading to significantly decreased ARID3A, ARID3B, and lysine demethylase 4C (KDM4C). Similarly, Sw.71 cells overexpressing LIN28A and LIN28B (DKIs) significantly decreased let-7 miRNAs, leading to significantly increased ARID3A, ARID3B, and KDM4C. In ACH-3P cells, ARID3A, ARID3B, and KDM4C make a triprotein complex [triprotein complex comprising ARID3A, ARID3B, and KDM4C (ARID3B-complex)] that binds the promoter regions of HMGA1, c-MYC, VEGF-A, and WNT1. ARID3B knockout in ACH-3P cells disrupted the ARID3B-complex, leading to a significant decrease in HMGA1, c-MYC, VEGF-A, and WNT1. DKOs had a significant reduction, whereas DKIs had a significant increase in HMGA1, c-MYC, VEGF-A, and WNT1, potentially due to regulation by the ARID3B-complex. This is the first study showing regulation of let-7 targets in immortalized human trophoblast cells by the ARID3B-complex.-Ali, A., Anthony, R. V., Bouma, G. J., Winger, Q. A. LIN28-let-7 axis regulates genes in immortalized human trophoblast cells by targeting the ARID3B-complex.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Regulación de la Expresión Génica , MicroARNs/fisiología , Proteínas de Unión al ARN/fisiología , Trofoblastos/metabolismo , Células Cultivadas , Femenino , Retardo del Crecimiento Fetal/metabolismo , Proteínas HMGA/genética , Humanos , Embarazo , Proteínas Proto-Oncogénicas c-myc/genética , Factores de Transcripción/fisiología , Factor A de Crecimiento Endotelial Vascular/genética , Proteína Wnt1/genética
13.
Life Sci ; 232: 116668, 2019 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-31326568

RESUMEN

Gene regulatory mechanisms determine the multistep carcinogenesis process. Two aspects of epigenetics are microRNA (miRNAs) and DNA methylation that regulate distinct biological mechanisms such as metastasis, apoptosis cell proliferation and induction of senescence. Although critical, the interplay between these two epigenetic mechanisms is yet to be completely understood, particularly in cervical cancer. To study the DNA methylation regulation of miRNAs and its potential role in cervical cancer, we investigated the differential methylation pattern of two candidate miRNAs (miR-375 and miR-196a-1) during cervical cancer progression against normal cervical epithelium (NCE) by bisulfite DNA sequencing. miR-375 and miR-196a-1 were hypermethylated in Squamous Cell Carcinoma (SCC) against NCE and Cervical Intra-Epithelial Neoplasia (CIN) (p < 0.05). Treatment with demethylating agent reactivated the miR-375 and miR-196a-1 expression in SiHa, HeLa and CaSki cells. In vitro artificial methylation by M.SssI followed by dual luciferase assay confirmed miR-375 and miR-196a-1 as methylation regulated miRNAs (P < 0.05). miR-375 and miR-196a-1 expression levels were negatively correlated with methylation levels in clinical specimens. We further identified Replication Factor C Subunit 3 (RFC3) and High Mobility Group AT-Hook 1 (HMGA1) as targets of miR-375 and miR-196a-1 respectively by dual luciferase reporter assay. Our analysis indicates that miR-375 and miR-196a-1 are DNA methylation regulated miRNAs whose deregulation may facilitate pathophysiology of cervical cancer.


Asunto(s)
Metilación de ADN , MicroARNs/genética , Neoplasias del Cuello Uterino/genética , Adulto , Anciano , Línea Celular Tumoral , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Proteínas HMGA/genética , Humanos , Persona de Mediana Edad , Regiones Promotoras Genéticas , Proteína de Replicación C/genética , Neoplasias del Cuello Uterino/patología
14.
Int J Mol Sci ; 20(11)2019 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-31167352

RESUMEN

Plasticity is an essential condition for cancer cells to invade surrounding tissues. The nucleus is the most rigid cellular organelle and it undergoes substantial deformations to get through environmental constrictions. Nuclear stiffness mostly depends on the nuclear lamina and chromatin, which in turn might be affected by nuclear architectural proteins. Among these is the HMGA1 (High Mobility Group A1) protein, a factor that plays a causal role in neoplastic transformation and that is able to disentangle heterochromatic domains by H1 displacement. Here we made use of atomic force microscopy to analyze the stiffness of breast cancer cellular models in which we modulated HMGA1 expression to investigate its role in regulating nuclear plasticity. Since histone H1 is the main modulator of chromatin structure and HMGA1 is a well-established histone H1 competitor, we correlated HMGA1 expression and cellular stiffness with histone H1 expression level, post-translational modifications, and nuclear distribution. Our results showed that HMGA1 expression level correlates with nuclear stiffness, is associated to histone H1 phosphorylation status, and alters both histone H1 chromatin distribution and expression. These data suggest that HMGA1 might promote chromatin relaxation through a histone H1-mediated mechanism strongly impacting on the invasiveness of cancer cells.


Asunto(s)
Neoplasias de la Mama/metabolismo , Núcleo Celular/metabolismo , Proteínas HMGA/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Ciclo Celular/genética , Línea Celular Tumoral , Cromatina/genética , Cromatina/metabolismo , Femenino , Expresión Génica , Proteínas HMGA/genética , Histonas/metabolismo , Humanos , Estimación de Kaplan-Meier , Fosforilación , Pronóstico , Unión Proteica
15.
Mol Med Rep ; 20(1): 584-592, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31115557

RESUMEN

The purpose of the present study was to investigate the differentially expressed proteins between endotoxin tolerance and sepsis. Cell models of an endotoxin tolerance group (ET group) and sepsis group [lipopolysaccharide (LPS) group] were established using LPS and evaluated using ELISA and flow cytometry methods. Differentially expressed proteins between the ET and the LPS groups were identified using isobaric tags for relative and absolute quantitation (iTRAQ) analysis and evaluated by bioinformatics analysis. The expression of core proteins was detected by western blotting. It was identified that the expression of tumor necrosis factor­α and interleukin­6 was significantly decreased in the ET group compared with the LPS group. Following high­dose LPS stimulation for 24 h, the positive rate of cluster of differentiation­16/32 in the ET group (79.07%) was lower when compared with that of the LPS group (94.27%; P<0.05). A total of 235 proteins were identified by iTRAQ, and 36 upregulated proteins with >1.2­fold differences and 27 downregulated proteins with <0.833­fold differences were detected between the ET and LPS groups. Furthermore, the expression of high mobility group (HMG)­A1 and HMGA2 in the ET group was higher compared with the LPS group following high­dose LPS stimulation for 4 h, while HMGB1 and HMGB2 exhibited the opposite expression trend under the same conditions. In conclusion, proteomics analysis using iTRAQ technology contributes to a deeper understanding of ET mechanisms. HMGA1, HMGA2, HMGB1 and HMGB2 may serve a crucial role in the development of ET.


Asunto(s)
Endotoxinas/farmacología , Proteínas HMGA/genética , Lipopolisacáridos/farmacología , Sepsis/tratamiento farmacológico , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular , Biología Computacional , Tolerancia a Medicamentos/genética , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Interleucina-6/genética , Proteómica , Sepsis/genética , Sepsis/patología , Factor de Necrosis Tumoral alfa/genética
16.
Int J Oncol ; 54(5): 1771-1784, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30816441

RESUMEN

Ovarian cancer (OC) is the gynecological malignancy type with the highest mortality rate in females. The regulatory effect of microRNAs (miRs) on their target genes serves a key role in tumor development. Therefore, in the present study, whether miR let­7d­5p targeting high mobility group A1 (HMGA1) regulated biological characteristics and chemosensitivity of OC cells by mediating the p53 signaling pathway was investigated. The let­7d­5p level was detected in OC tissues and adjacent normal tissues, followed by detection in OC cell lines SKOV3, A2780, OVCAR­3 and CaOV3, and human normal ovarian epithelial cell line (IOSE­80), in order to select the OC cell line for the following experiments. Subsequently, OC cells were treated with the let­7d­5p mimic, siHMGA1 and Tenovin­1. The targeting association between let­7d­5p and HMGA1 was then examined, and the OC cell viability, migration, cycle and apoptosis were evaluated. Subsequently, the chemosensitivity of OC cells to cisplatin was verified. Finally, expression levels of let­7d­5p, HMGA1, p21, B­cell lymphoma­2 (Bcl­2)­associated X (Bax), p27, p53 wild­type (p53wt), p53 mutated (p53mut), proliferating cell nuclear antigen (PCNA), cyclin­dependent kinase 2 (CDK2), matrix metallopeptidase (MMP)2, MMP9 and Bcl­2 were determined. As demonstrated in the results, let­7d­5p expression was low in OC tissues and had an increased reduction in the OVCAR­3 cell line. HMGA1 was confirmed as a target of let­7d­5p, and its expression was also silenced by let­7d­5p. let­7d­5p repressed OC cell viability, migration, cell cycle progression and apoptosis, while it promoted the chemosensitivity of OC cells to cisplatin by targeting HMGA1. The expression of let­7d­5p, p21, Bax, p27 and p53wt was increased, while that of HMGA1, p53mut, PCNA, CDK2, MMP2, MMP9 and Bcl­2 was reduced following cell transfection. The results in the present study provided evidence that let­7d­5p may suppress proliferation, and facilitate apoptosis and cisplatin chemosensitivity of OC cells by silencing HMGA1 via the p53 signaling pathway.


Asunto(s)
Regulación hacia Abajo , Resistencia a Antineoplásicos , Proteínas HMGA/genética , MicroARNs/genética , Neoplasias Ováricas/genética , Transducción de Señal , Adulto , Línea Celular Tumoral , Cisplatino/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , MicroARNs/farmacología , Persona de Mediana Edad , Transducción de Señal/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo
17.
Eur Rev Med Pharmacol Sci ; 23(2): 740-748, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30720182

RESUMEN

OBJECTIVE: Dysfunction of the microRNA (miRNA) network is a major regulator in neurological diseases. However, little is known about the functional significance of miRs in ischemic brain injury. This study was designed to investigate the functional behaviors and regulatory mechanisms of miR-196a in ischemic brain injury. MATERIALS AND METHODS: Cerebral ischemia was induced by middle cerebral artery occlusion (MCAO) in rats. The expression levels of miR-196a and HMGA1 were detected by quantitative Real Time-Polymerase Chain Reaction (qRT-PCR), Western blotting, and/or immunocytochemistry. The role of miR-196a in cerebral infarction and brain cell apoptosis was determined by infarct volume estimation and Transferase-mediated deoxyuridine triphosphate-biotin nick end labeling (TUNEL) assay, respectively. Bioinformatics software and Luciferase analysis were used to predict and confirm the targets of miR-196a. RESULTS: The results showed that the miR-196a expression was significantly increased in rat brain after MCAO and in cultured neonatal rat cortical neurons after oxygen-glucose deprivation (OGD). Pretreatment with antagomiR-196a by intracerebroventricular markedly reduced the miR-196a mRNA and HMGA1 protein of the brain in rats subjected to MCAO. Notably, the knockdown of miR-196a could protect MCAO rats against cerebral infarction and brain cell apoptosis. In the OGD model, apoptosis and miR-196a expressions were inhibited, while High-Mobility-Group-A1 (HMGA1) levels were increased in cortical neurons in a time-dependent manner. Moreover, HMGA1 was the target gene of miR-196a. MiR-196a overexpression promoted OGD-induced cortical neurons apoptosis possibly through negative regulation of HMGA1. CONCLUSIONS: These findings indicated a crucial role of miR-196a in regulating infarct volume and neuronal cell death under cerebral ischemia, thus offering a new target for the development of therapeutic agents against ischemic brain injury.


Asunto(s)
Corteza Cerebral/patología , Proteínas HMGA/genética , Proteína HMGA1a/genética , Infarto de la Arteria Cerebral Media/genética , MicroARNs/metabolismo , Animales , Apoptosis/genética , Hipoxia de la Célula , Células Cultivadas , Corteza Cerebral/citología , Modelos Animales de Enfermedad , Regulación hacia Abajo , Técnicas de Silenciamiento del Gen , Humanos , Infarto de la Arteria Cerebral Media/patología , Masculino , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Neuronas/patología , Cultivo Primario de Células , Ratas
18.
Biomed Pharmacother ; 108: 43-49, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30216798

RESUMEN

Diffuse large B-cell lymphoma (DLBCL) is the most common subtype of non-Hodgkin lymphoma, which is an aggressive malignancy with high variance of clinical features and response to the treatment. The proteasome inhibitor bortezomib (BTZ) has been demonstrated to suppress the progression of DLBCL, however, the underlying molecular mechanisms by which BTZ regulates the growth of DLBCL cells remain largely unknown. Increasing evidence has suggested that microRNAs (miRNAs) are novel targets of anti-cancer drugs to modulate the progression of cancers. Here, we showed BTZ treatment significantly inhibited the proliferation of DLBCL CRL-2630 cells. Mechanistically, exposure of BTZ up-regulated the expression of miR-198 in DLBCL cells. Depletion of miR-198 significantly reversed the inhibitory effect of BTZ on the proliferation of CRL-2630 cells. To further characterize the involvement of miR-198 in BTZ-induced growth defects of CRL-2630 cells, the downstream targets of miR-198 were predicted with the bioinformatics tools. The results showed that miR-198 bound the 3'-untranslated region (UTR) of the high mobility group AT-hook 1 (HMGA1) and suppressed the expression of HMGA1 in DLBCL cells. Consistently, BTZ treatment decreased the level of HMAG1 and inhibited the migration of DLBCL cells. Our results provided the possible mechanism by which BTZ suppressed the growth of DLBCL cells.


Asunto(s)
Bortezomib/farmacología , Progresión de la Enfermedad , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/patología , MicroARNs/metabolismo , Secuencia de Bases , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas HMGA/genética , Proteínas HMGA/metabolismo , Humanos , MicroARNs/genética , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
19.
Clin Cancer Res ; 24(24): 6367-6382, 2018 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-30135148

RESUMEN

PURPOSE: The study of the cancer secretome suggests that a fraction of the intracellular proteome could play unanticipated roles in the extracellular space during tumorigenesis. A project aimed at investigating the invasive secretome led us to study the alternative extracellular function of the nuclear protein high mobility group A1 (HMGA1) in breast cancer invasion and metastasis. EXPERIMENTAL DESIGN: Antibodies against HMGA1 were tested in signaling, adhesion, migration, invasion, and metastasis assays using breast cancer cell lines and xenograft models. Fluorescence microscopy was used to determine the subcellular localization of HMGA1 in cell lines, xenograft, and patient-derived xenograft models. A cohort of triple-negative breast cancer (TNBC) patients was used to study the correlation between subcellular localization of HMGA1 and the incidence of metastasis. RESULTS: Our data show that treatment of invasive cells with HMGA1-blocking antibodies in the extracellular space impairs their migration and invasion abilities. We also prove that extracellular HMGA1 (eHMGA1) becomes a ligand for the Advanced glycosylation end product-specific receptor (RAGE), inducing pERK signaling and increasing migration and invasion. Using the cytoplasmic localization of HMGA1 as a surrogate marker of secretion, we showed that eHMGA1 correlates with the incidence of metastasis in a cohort of TNBC patients. Furthermore, we show that HMGA1 is enriched in the cytoplasm of tumor cells at the invasive front of primary tumors and in metastatic lesions in xenograft models. CONCLUSIONS: Our results strongly suggest that eHMGA1 could become a novel drug target in metastatic TNBC and a biomarker predicting the onset of distant metastasis.


Asunto(s)
Proteínas HMGA/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Espacio Extracelular/metabolismo , Femenino , Expresión Génica , Proteínas HMGA/genética , Proteína HMGA1a/metabolismo , Xenoinjertos , Humanos , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia , Estadificación de Neoplasias , Fenotipo , Unión Proteica , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Transducción de Señal , Neoplasias de la Mama Triple Negativas/genética
20.
Mol Cell Biol ; 38(19)2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29987187

RESUMEN

Peptidylarginine deiminase (PAD) enzymes convert histone arginine residues into citrulline to modulate chromatin organization and gene expression. Although PADs are expressed in anterior pituitary gland cells, their functional role and expression in pituitary adenomas are unknown. To begin to address these issues, we first examined normal human pituitaries and pituitary adenomas and found that PAD2, PAD4, and citrullinated histones are highest in prolactinomas and somatoprolactinomas. In the somatoprolactinoma-derived GH3 cell line, PADs citrullinate histone H3, which is attenuated by a pan-PAD inhibitor. RNA sequencing and chromatin immunoprecipitation (ChIP) studies show that the expression of microRNAs (miRNAs) let-7c-2, 23b, and 29c is suppressed by histone citrullination. Our studies demonstrate that these miRNAs directly target the mRNA of the oncogenes encoding HMGA, insulin-like growth factor 1 (IGF-1), and N-MYC, which are highly implicated in human prolactinoma/somatoprolactinoma pathogenesis. Our results are the first to define a direct role for PAD-catalyzed histone citrullination in miRNA expression, which may underlie the etiology of prolactinoma and somatoprolactinoma tumors through regulation of oncogene expression.


Asunto(s)
Histonas/metabolismo , Lactotrofos/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Oncogenes , ARN Mensajero/genética , ARN Mensajero/metabolismo , Somatotrofos/metabolismo , Células Cultivadas , Citrulinación/genética , Epigénesis Genética , Expresión Génica , Técnicas de Silenciamiento del Gen , Proteínas HMGA/genética , Proteínas HMGA/metabolismo , Código de Histonas/genética , Histonas/química , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Lactotrofos/citología , Modelos Biológicos , Proteína Proto-Oncogénica N-Myc/genética , Proteína Proto-Oncogénica N-Myc/metabolismo , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/metabolismo , Prolactinoma/genética , Prolactinoma/metabolismo , Arginina Deiminasa Proteína-Tipo 2 , Arginina Deiminasa Proteína-Tipo 4 , Desiminasas de la Arginina Proteica/antagonistas & inhibidores , Desiminasas de la Arginina Proteica/genética , Desiminasas de la Arginina Proteica/metabolismo , Somatotrofos/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...