Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(14)2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-39062899

RESUMEN

HMGB3 protein belongs to the group of HMGB proteins from the superfamily of nuclear proteins with high electrophoretic mobility. HMGB proteins play an active part in almost all cellular processes associated with DNA-repair, replication, recombination, and transcription-and, additionally, can act as cytokines during infectious processes, inflammatory responses, and injuries. Although the structure and functions of HMGB1 and HMGB2 proteins have been intensively studied for decades, very little attention has been paid to HMGB3 until recently. In this review, we summarize the currently available data on the molecular structure, post-translational modifications, and biological functions of HMGB3, as well as the possible role of the ubiquitin-proteasome system-dependent HMGB3 degradation in tumor development.


Asunto(s)
Proteína HMGB3 , Procesamiento Proteico-Postraduccional , Humanos , Proteína HMGB3/metabolismo , Proteína HMGB3/química , Proteína HMGB3/genética , Animales , Neoplasias/metabolismo , Proteolisis , Complejo de la Endopetidasa Proteasomal/metabolismo
2.
Tissue Cell ; 88: 102406, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38761792

RESUMEN

BACKGROUND: Previous evidences has highlighted the pivotal role of NOD-like receptor family pyrin domain-containing 3 (NLRP3)-mediated inflammasomes and pyroptosis activation in driving tumor malignancy and shaping the tumor microenvironment. Herein, we aimed to elucidate the impact of high-mobility group box 3 (HMGB3) released in glioma-derived exosomes on macrophage infiltration in gliomas, NLRP3 inflammasome activation and polarization. METHODS: Transcripts and protein levels of HMGB3, and cytokines associated with macrophage phenotypes and pyroptosis were assessed in glioma tissues and cell lines (U251, LN229, T98G, A172) using qRT-PCR and/or Western blot analysis. Exosomes secreted from LN229 and NHA cells were isolated via differential ultracentrifugation and characterized by transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA), and analysis of exosome-related markers. PKH67 staining was employed to examine exosomes uptake by THP-1 differentiated macrophages. Flow cytometry was utilized to assess macrophage pyroptotic rates and polarization-related markers. RESULTS: HMGB3 expression was elevated in glioma tissues, serum samples and tumor cell lines. Kaplan-Meier curves revealed a positive correlation between higher HMGB3 expression and poor overall survival and recurrence-free survival. Moreover, glioma tissues with increased HMGB3 expression exhibited significant upregulation of M2 macrophages markers (CD68, CD206, Arg1) and NLRP3 inflammasome components (NLRP3, IL-1ß, ASC), suggesting that HMGB3 was closely associated with macrophage infiltration and NLRP3 inflammasome activation. Notably, HMGB3 was found to be enriched in glioma cell- secreted exosomes and could be internalized by macrophages. Knockdown of HMGB3 in glioma cell exosomes could restrain M2 macrophage polarization, NLRP3 inflammasome activation and pyroptosis. CONCLUSION: These findings suggested that glioma cells secreted exosomal HMGB3 could facilitate macrophage M2 polarization, pyroptosis and inflammatory infiltration, indicating HMGB3 might be a poor prognosis factor for glioma.


Asunto(s)
Exosomas , Glioma , Proteína HMGB3 , Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Piroptosis , Macrófagos Asociados a Tumores , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Exosomas/metabolismo , Glioma/patología , Glioma/metabolismo , Glioma/genética , Humanos , Inflamasomas/metabolismo , Línea Celular Tumoral , Proteína HMGB3/metabolismo , Proteína HMGB3/genética , Macrófagos Asociados a Tumores/metabolismo , Macrófagos Asociados a Tumores/patología , Masculino , Femenino , Microambiente Tumoral , Macrófagos/metabolismo , Macrófagos/patología , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/genética
3.
Cell Commun Signal ; 21(1): 144, 2023 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-37328851

RESUMEN

BACKGROUND: Ovarian cancer, particularly epithelial ovarian cancer (EOC), is the leading cause of cancer-related mortality among women. Our previous study revealed that high HMGB3 levels are associated with poor prognosis and lymph node metastasis in patients with high-grade serous ovarian carcinoma; however, the role of HMGB3 in EOC proliferation and metastasis remains unknown. METHODS: MTT, clonogenic, and EdU assays were used to assess cell proliferation. Transwell assays were performed to detect cell migration and invasion. Signaling pathways involved in HMGB3 function were identified by RNA sequencing (RNA-seq). MAPK/ERK signaling pathway protein levels were evaluated by western blot. RESULTS: HMGB3 knockdown inhibited ovarian cancer cell proliferation and metastasis, whereas HMGB3 overexpression facilitated these processes. RNA-seq showed that HMGB3 participates in regulating stem cell pluripotency and the MAPK signaling pathway. We further proved that HMGB3 promotes ovarian cancer stemness, proliferation, and metastasis through activating the MAPK/ERK signaling pathway. In addition, we demonstrated that HMGB3 promotes tumor growth in a xenograft model via MAPK/ERK signaling. CONCLUSIONS: HMGB3 promotes ovarian cancer malignant phenotypes and stemness through the MAPK/ERK signaling pathway. Targeting HMGB3 is a promising strategy for ovarian cancer treatment that may improve the prognosis of women with this disease. Video Abstract.


Asunto(s)
Proteína HMGB3 , Neoplasias Ováricas , Transducción de Señal , Femenino , Humanos , Carcinoma Epitelial de Ovario/genética , Carcinoma Epitelial de Ovario/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Ováricas/patología , Fenotipo , Proteína HMGB3/genética
4.
Biochem Genet ; 61(3): 1210-1229, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36542208

RESUMEN

The aim of this study was to investigate the role of circRNA insulin growth factor 1 receptor (circ-IGF1R) in colorectal cancer (CRC). Glycolytic metabolism was analyzed by glucose uptake and lactate production using the corresponding kits. The protein levels were determined using Western blot. The effect of circ-IGF1R on CRC in vivo was explored by xenograft experiment in mice. Circ-IGF1R was up-regulated in CRC tissues and cells. Knockdown of circ-IGF1R inhibited proliferation, migration, invasion and glycolysis but induced apoptosis of CRC cells. Circ-IGF1R interacted with miR-362-5p and miR-362-5p inhibitor attenuated the anti-tumor effects of circ-IGF1R downregulation on CRC cells. HMGB3 acted as a downstream target for miR-362-5p, and circ-IGF1R facilitated the malignant behaviors of CRC cells by regulating HMGB3. Circ-IGF1R activated the Wnt/ß-catenin pathway via targeting miR-362-5p/HMGB3 axis. Tumor growth in vivo was reduced after knockdown of circ-IGF1R. Circ-IGF1R might be a novel biomolecular target for CRC diagnosis and treatment.


Asunto(s)
Neoplasias Colorrectales , MicroARNs , ARN Circular , Animales , Humanos , Ratones , beta Catenina/genética , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/genética , Ácido Láctico , MicroARNs/genética , Receptor IGF Tipo 1/genética , Transducción de Señal , Factores de Transcripción , ARN Circular/genética , ARN Circular/metabolismo , Proteína HMGB3/genética , Proteína HMGB3/metabolismo
5.
Mol Cell Biochem ; 478(9): 1973-1986, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-36586093

RESUMEN

Tanshinone IIA (TSIIA), a multi-pharmaceutical compound, has been demonstrated to have anti-tumor properties. This study explores the potential regulatory mechanism of TSIIA on non-small cell lung cancer (NSCLC) progression. The cytotoxicity of TSIIA was evaluated by MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide) and LDH (lactate dehydrogenase) assays. Expression levels of circ_0020123 (hsa_circ_0020123) and microRNA-1299 (miR-1299) were assessed by quantitative real-time polymerase chain reaction (qRT-PCR). Cell proliferation, migration, invasion, and apoptosis were analyzed by MTT, colony formation, transwell, wound-healing, or flow cytometry assays. The relationship between miR-1299 and circ_0020123 or HMGB3 (high mobility group box 3) was verified by the dual-luciferase reporter and/or RNA immunoprecipitation (RIP) assays. Protein level of HMGB3 was measured by western blotting. The relationship between TSIIA and circ_0020123 was confirmed by xenograft assay. TSIIA reduced xenograft tumor growth in vivo and repressed proliferation, migration, invasion, and facilitated apoptosis of NSCLC cells in vitro. TSIIA reduced circ_0020123 and HMGB3 expression, whereas elevated miR-1299 expression in NSCLC cells. Circ_0020123 knockdown enhanced the repressive influence of TSIIA treatment on the malignancy of NSCLC cells in vitro and in vivo. Circ_0020123 sponged miR-1299 to regulate HMGB3 expression under TSIIA treatment. MiR-1299 inhibitor reversed circ_0020123 knockdown-mediated influence on malignant behaviors of NSCLC cells under TSIIA treatment. HMGB3 elevation offset the suppressive impact of miR-1299 mimic on the malignancy of NSCLC cells under TSIIA treatment. TSIIA curbed NSCLC progression by the circ_0020123/miR-1299/HMGB3 axis, manifesting that the TSIIA/circ_0020123/miR-1299/HMG regulatory network might be a potential treatment strategy for NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Proteína HMGB3 , Neoplasias Pulmonares , MicroARNs , Humanos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Abietanos/farmacología , Proliferación Celular , Proteína HMGB3/genética , MicroARNs/genética
6.
Prostate ; 83(5): 440-453, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36541373

RESUMEN

BACKGROUND: The homeodomain-containing transcription factor NANOG is overexpressed in prostate adenocarcinoma (PCa) and predicts poor prognosis. The SOX family transcription factor SOX9, as well as the transcription co-activator HMGB3 of the HMGB family, are also overexpressed and may play pivotal roles in PCa. However, it is unknown whether SOX9 and HMGB3 interact with each other, or if they regulate NANOG gene transcription. METHODS: We identified potential SOX9 responsive elements in NANOG promoter, and investigated if SOX9 regulated NANOG transcription in co-operation with HMGB3 by experimental analysis of potential SOX9 binding sites in NANOG promoter, reporter gene transcription assays with or without interference or artificial overexpression of SOX9 and/or HMGB3, and protein-binding assays of SOX9-HMGB3 interaction. Clinicopathologic and prognostic significance of SOX9-HMGB3 overexpression in PCa was analyzed. RESULTS: SOX9 activated NANOG gene transcription by preferentially binding to a highly conserved consensus cis-regulatory element (-573 to -568) in NANOG promoter, and promoted the expression of NANOG downstream oncogenic genes. Importantly, HMGB3 functioned as a partner of SOX9 to co-operatively enhance transactivation of NANOG by interacting with SOX9, predominantly via the HMG Box A domain of HMGB3. Overexpression of SOX9 and/or HMGB3 enhanced PCa cell survival and cell migration and were significantly associated with PCa progression. Notably, Cox proportional regression analysis showed that co-overexpression of both SOX9 and HMGB3 was an independent unfavorable prognosticator for both CRPC-free survival (relative risk [RR] = 3.779,95% confidence interval [CI]: 1.159-12.322, p = 0.028) and overall survival (RR = 3.615,95% CI: 1.101-11.876, p = 0.034). CONCLUSIONS: These findings showed a novel SOX9/HMGB3/NANOG regulatory mechanism, deregulation of which played important roles in PCa progression.


Asunto(s)
Proteína HMGB3 , Proteína Homeótica Nanog , Neoplasias de la Próstata , Factor de Transcripción SOX9 , Humanos , Masculino , Regulación de la Expresión Génica , Proteína HMGB3/genética , Proteína HMGB3/metabolismo , Proteína Homeótica Nanog/genética , Proteína Homeótica Nanog/metabolismo , Procesos Neoplásicos , Próstata/metabolismo , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Factores de Transcripción/genética
7.
Cell Cycle ; 22(23-24): 2584-2601, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38197217

RESUMEN

The family of high mobility group box (HMGB) proteins participates in various biological processes including immunity, inflammation, as well as cancer formation and progression. However, its role in thyroid cancer remains to be clarified. We performed quantitative RT-PCR (qRT-PCR), western blot, enzyme-linked immunosorbent, immunohistochemistry, and immunofluorescence assays to evaluate the expression level and subcellular location of HMGB3. The effects of HMGB3 knockdown on malignant biological behaviors of thyroid cancer were determined by cell proliferation assays, cell cycle and apoptosis assays, and transwell chamber migration and invasion assays. Differential expression genes (DEGs) altered by HMGB3 were analyzed using the Ingenuity Pathway Analysis (IPA) and TRRUST v2 database. HMGB3 correlated pathways predicted by bioinformatic analysis were then confirmed using western blot, co-immunoprecipitation, dual-luciferase reporter assay, and flow cytometry. We found that HMGB3 is overexpressed and its downregulation inhibits cell viability, promotes cell apoptosis and cell cycle arrest, and suppresses cell migration and invasion in thyroid cancer. In PTC, both tissue and serum levels of HMGB3 are elevated and are correlated with lymph node metastasis and advanced tumor stage. Mechanistically, we observed the translocation of HMGB3 in PTC, induced at least partially by hypoxia. Cytoplasmic HMGB3 activates nucleic-acid-mediated TLR3/NF-κB signaling and extracellular HMGB3 interacts with the transmembrane TREM1 receptor in PTC. This study demonstrates the oncogenic role of HMGB3 cytoplasmic and extracellular translocation in papillary thyroid cancers; we recommend its future use as a potential circulating biomarker and therapeutic target for PTC.


Asunto(s)
Proteína HMGB3 , MicroARNs , Neoplasias de la Tiroides , Humanos , Cáncer Papilar Tiroideo/genética , Cáncer Papilar Tiroideo/patología , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/metabolismo , Línea Celular Tumoral , Receptor Activador Expresado en Células Mieloides 1/genética , Receptor Activador Expresado en Células Mieloides 1/metabolismo , Neoplasias de la Tiroides/genética , Proteína HMGB3/genética , Proteína HMGB3/metabolismo , Proliferación Celular/genética , MicroARNs/genética , Movimiento Celular/genética , Regulación Neoplásica de la Expresión Génica
8.
Life Sci ; 304: 120727, 2022 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-35753437

RESUMEN

AIMS: High mobility group box (HMGB) family proteins, HMGB1, HMGB2, HMGB3, and HMGB4 are oncogenic. The oncogenic nature of HMGB1 is characterized by its association with autophagy, ROS, and MMP. Since HMGB3 is its paralog, we hypothesized that it might also modulate autophagy, ROS, and MMP. Hence, we targeted HMGB3 using its shRNA or miR-142-3p and assessed the changes in autophagy, ROS, MMP, and tumorigenic properties of human breast cancer cells. MAIN METHODS: Cell viability was assessed by resazurin staining and annexin-V/PI dual staining was used for confirming apoptosis. Colony formation, transwell migration, invasion and luciferase reporter (for miRNA-target validation) assays were also performed. ROS and MMP were detected using DHE and MitoTracker dyes, respectively. A zebrafish xenograft model was used to assess the role of miR-142-3p on in vivo metastatic potential of breast cancer cells. KEY FINDINGS: Breast cancer tissues from Indian patients and TCGA samples exhibit overexpression of HMGB3. miR-142-3p binds to 3' UTR of HMGB3, leading to its downregulation that subsequently inhibits colony formation and induces apoptosis involving increased ROS accumulation and decreased MMP, phospho-mTOR and STAT3. Our findings show that HMGB3 is directly involved in the miR-142-3p-mediated disruption of autophagy and induction of apoptotic cell death via modulation of LC3, cleaved PARP and Bcl-xL. In addition, miR-142-3p inhibited migration, invasion and metastatic potential of breast cancer cells. SIGNIFICANCE: Our findings highlighted the role of HMGB3, for the first time, in the modulation of autophagy and apoptosis in human breast cancer cells, and these results have therapeutic implications.


Asunto(s)
Neoplasias de la Mama , Proteína HMGB1 , Proteína HMGB3 , MicroARNs , Regiones no Traducidas 3' , Animales , Apoptosis/genética , Autofagia , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular/genética , Femenino , Proteína HMGB1/genética , Proteína HMGB3/genética , Proteína HMGB3/metabolismo , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno , Pez Cebra/genética
9.
Gynecol Obstet Invest ; 87(3-4): 200-210, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35705019

RESUMEN

OBJECTIVES: The aim of the study was to find out the function of long noncoding RNA brain cytoplasmic RNA 1 (BCYRN1) in cisplatin (DDP)-resistance of cervical cancer (CC) cells. Design and Materials, Setting, Methods: BCYRN1 expression in CC and DDP-resistant cells was evaluated, with the association of BCYRN1 and prognosis analyzed. Then, DDP-resistant cells with BCYRN1 knockdown were established and the DDP-resistance of these cells was assessed. BCYRN1 subcellular localization was detected and confirmed. Besides, the binding relations of BCYRN1 and microRNA (miR)-330-5p and between miR-330-5p and high-mobility group box 3 (HMGB3) were examined and verified. Moreover, the role of miR-330-5p and HMGB3 in the mechanism of BCYRN1 modulating DDP-resistance of CC cells was detected. In addition, xenograft transplantation was conducted to confirm the effect of BCYRN1 in CC cell DDP-resistance. RESULTS: BCYRN1 was overexpressed in CC, which resulted in poor prognosis and DDP-resistance. BCYRN1 knockdown in DDP-resistant cells downregulated DDP-resistance. Mechanically, BCYRN1 sponged miR-330-5p to strengthen HMGB3 mRNA level. Besides, miR-330-5p underexpression or HMGB3 overexpression reversed the function of BCYRN1 knockdown in inhibiting DDP-resistance of CC cells. Eventually, BCYRN1 knockdown reduced the DDP-resistance of CC cells in vivo. LIMITATIONS: There are still some deficiencies in the research; for example, whether there are other miRs working as the downstream genes of BCYRN1 in the competing endogenous RNA interaction is not fully clarified, nor the other downstream mechanisms of miR-330-5p. Besides, the experimental findings and their application into practice need extensive validation. CONCLUSIONS: BCYRN1 knockdown could disrupt the DDP-resistance of CC cells through upregulating miR-330-5p to suppress HMGB3 mRNA level.


Asunto(s)
Resistencia a Antineoplásicos , Proteína HMGB3 , MicroARNs , ARN Largo no Codificante , Neoplasias del Cuello Uterino , Encéfalo/metabolismo , Línea Celular Tumoral , Proliferación Celular , Cisplatino/farmacología , Resistencia a Antineoplásicos/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Proteína HMGB3/genética , Humanos , MicroARNs/genética , ARN Largo no Codificante/genética , ARN Mensajero , Neoplasias del Cuello Uterino/tratamiento farmacológico , Neoplasias del Cuello Uterino/genética
10.
Bioengineered ; 13(4): 9959-9972, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35416122

RESUMEN

The recurrence and metastasis of gastric cancer are related to the stemness of gastric cancer cells. Researches have shown that miR-18 level is negatively correlated to the occurrence and development of certain cancer types. However, the effects of miR-18 on the stemness of gastric cancer remain uncertain. In this research, gastric cancer cell lines with stable overexpression of miR-18 were constructed through lentivirus infection. CCK-8 assay, RT-qPCR, Western blot, flow cytometry, and in vivo tumorigenesis assays were performed to evaluate the effects of miR-18 on the stemness of gastric cancer cells. Moreover, luciferase reporter assays found that Meis2 was the target of miR-18. Furthermore, we also found that the low-expressed oncogene HMGB3 is involved in this miR-18/Meis2 axis to further promote the stemness of gastric cancer cells. These findings suggest that the miR-18/Meis2/HMGB3 axis may be potential prognostic indicators for patients with gastric cancer.


Asunto(s)
Proteína HMGB3 , MicroARNs , Neoplasias Gástricas , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Proteína HMGB3/genética , Proteína HMGB3/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Oncogenes , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Factores de Transcripción/metabolismo
11.
Endocr Relat Cancer ; 29(4): 175-189, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35073278

RESUMEN

Thyroid cancer is one of the most common endocrine malignancies. It is necessary to discover more effective molecular targets for the treatment of thyroid cancer. The results of immunohistochemical staining, qPCR and Western blot indicated that the expression of SYT7 in thyroid cancer tissues and cells was higher than that in paracarcinoma tissues and normal thyroid cells. Through cell function testing experiments, it was found that SYT7 knockdown inhibited the proliferation and migration of thyroid cancer cells and promoted cell apoptosis, while SYT7 overexpression had the opposite effect. Similarly, SYT7 downregulation also suppressed tumor growth in vivo. HMGB3 was confirmed to be the downstream gene of SYT7 by GeneChip and Ingenuity Pathway Analysis. Besides, through UbiBrowser database predictions and Co-IP assays, we found that SYT7 interacted with BRCA1 to inhibit HMGB3 ubiquitination and thus upregulated the protein level of HMGB3. Similar to SYT7, HMGB3 was significantly upregulated in thyroid cancer. HMGB3 knockdown inhibited the proliferation and migration of thyroid cancer cells and promoted cell apoptosis. Furthermore, HMGB3 knockdown restored the promotion of cell proliferation and migration caused by SYT7 overexpression. SYT7 and HMGB3 were upregulated in thyroid cancer, and SYT7 regulated the expression of HMGB3 through BRCA1-mediated ubiquitination of HMGB3 to promote thyroid cancer progression.


Asunto(s)
Proteína HMGB3 , MicroARNs , Neoplasias de la Tiroides , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Proteína HMGB3/genética , Proteína HMGB3/metabolismo , Humanos , MicroARNs/genética , Sinaptotagminas/genética , Sinaptotagminas/metabolismo , Neoplasias de la Tiroides/genética , Ubiquitinación
12.
Life Sci ; 291: 120268, 2022 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-34973275

RESUMEN

AIMS: High-mobility group (HMG) proteins are oncogenic in different cancers, including cervical cancer; silencing their individual expression using sh-RNAs, siRNAs, and miRNAs has had anti-tumorigenic effects, but the consequences of their collective downregulation are not known. Since multiple gene targeting is generally very effective in cancer therapy, the present study highlighted the consequences of silencing the expression of HMGA1, A2, B1, and B3 using sh-RNAs or miR-142-3p (that can potentially target HMGA1, A2, B1, and B3) in cervical cancer cell lines. MAIN METHODS: 3' UTR luciferase reporter assays were performed to validate HMGA1, A2, B1, and B3 as targets of miR-142-3p in human cervical cancer cells. Annexin V/PI dual staining and flow cytometry analyses were used to detect apoptotic cells. miR-142-3p-mediated regulation of cell death, colony formation, migration, and invasion was investigated in human cervical cancer cells together with in vivo metastasis in zebrafish. KEY FINDINGS: Concurrent knockdown of HMGA1, A2, B1, and B3 through their corresponding sh-RNAs inhibited cell viability and colony formation but induced apoptosis, and these effects were relatively reduced upon their individual knockdown. miR-142-3p targeted HMGA1, A2, B1, and B3 by binding to their 3'UTRs and induced apoptosis but inhibited proliferation, migration, and invasion of human cervical cancer cells. In addition, miR-142-3p expression decreased phospho-p65 and EMT-related proteins in cervical cancer cells and their in vivo metastatic potential upon implantation in zebrafish. SIGNIFICANCE: These findings suggest that miR-142-3p acts as a tumor-suppressive miRNA by targeting HMGA1, A2, B1, and B3 and may serve as a potential therapeutic agent in human cervical cancer.


Asunto(s)
MicroARNs/genética , Neoplasias del Cuello Uterino/metabolismo , Animales , Carcinogénesis/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Femenino , Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/genética , Proteína HMGA1a/genética , Proteína HMGA1a/metabolismo , Proteína HMGA2/genética , Proteína HMGA2/metabolismo , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Proteína HMGB3/genética , Proteína HMGB3/metabolismo , Células HeLa , Humanos , MicroARNs/metabolismo , Modelos Animales , Invasividad Neoplásica/genética , Oncogenes , Neoplasias del Cuello Uterino/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Pez Cebra
13.
Bioengineered ; 12(2): 12107-12122, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34753396

RESUMEN

Colorectal cancer (CRC) is the third most commonly diagnosed malignant tumor worldwide. LINC00857 has been reported as a dysregulated long non-coding RNAs (lncRNAs) involved in the genesis and development of different cancers. In CRC, accumulating evidence indicates that high mobility group box 3 (HMGB3) is over-expressed and contributes to CRC development. However, the mechanism underlying HMGB3 upregulation in CRC remains unclear. The present work aims to investigate the role of LINC00857 and its functional interaction with HMGB3 in regulating CRC progression. Differential expression of LINC00857 between CRC tissues and normal tissues was identified in TCGA (The Cancer Genome Atlas) database. In vitro functional assays were performed to explore the biological functions of LINC00857 in CRC cells. In vivo xenograft model was employed to investigate the role of LINC00857 in CRC tumorigenesis. We found that LINC00857 was significant upregulated in CRC tissues and cell lines. LINC00857 knockdown significantly inhibited the proliferation, migration and invasion of CRC cells, and also induced apoptosis. Moreover, LINC00857 knockdown suppressed CRC tumorigenesis in vivo. We further demonstrated that the effects of LINC00857 in CRC cells were mediated through miR-150-5p/HMGB3 axis. LINC00857 negatively regulates the activity of miR-150-5p, which releases its inhibition on HMGB3 expression. Our data indicate that LINC00857/miR-150-5p/HMGB3 axis plays a fundamental role in regulating the malignant phenotype and tumorigenesis of CRC. Targeting this axis may serve as novel therapeutic strategies for CRC treatment.


Asunto(s)
Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Proteína HMGB3/genética , MicroARNs/metabolismo , ARN Largo no Codificante/metabolismo , Regulación hacia Arriba/genética , Animales , Apoptosis/genética , Secuencia de Bases , Carcinogénesis/genética , Carcinogénesis/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Femenino , Técnicas de Silenciamiento del Gen , Proteína HMGB3/metabolismo , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/genética , Persona de Mediana Edad , Invasividad Neoplásica , ARN Largo no Codificante/genética
14.
Exp Mol Med ; 53(8): 1218-1228, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34408262

RESUMEN

Laryngeal squamous cell carcinoma (LSCC) is one of the most common subtypes of head and neck malignancies worldwide. Long intervening/intergenic noncoding RNAs (LINCRNAs) have been recently implicated in various biological processes that take place in the setting of laryngeal cancer, but the regulatory role of LINC00319 in LSCC remains largely unknown. The current study aimed to elucidate the regulatory effect of LINC00319 on the development and progression of LSCC via high-mobility group box 3 (HMGB3). Microarray-based analysis was initially conducted to identify differentially expressed long noncoding RNAs, after which the expression of LINC00319 and HMGB3 in LSCC tissues and cells was determined accordingly. CD133+CD144+ TU177 cells were subsequently isolated and transfected with LINC00319 overexpression vector (oe-LINC00319), short hairpin RNA (sh)-LINC00319, sh-HMGB3, sh-E2F transcription factor 1 (E2F1), and oe-E2F1, as well as their corresponding controls. The proliferative, invasion, self-renewal, and tumorigenic abilities of CD133+CD144+ TU177 cells were then evaluated. Our in vitro findings were further confirmed following subcutaneous injection of cells expressing the corresponding plasmids into nude mice. LINC00319 and HMGB3 expressions were elevated in LSCC cells and tissues. LINC00319 increased HMGB3 expression by recruiting E2F1. Furthermore, the stimulatory role of LINC00319 on the proliferation, invasion, self-renewal ability, and tumorigenicity of CD133+CD144+ TU177 cells was achieved by upregulating HMGB3 via recruitment of E2F1. The in vitro findings were also confirmed by in vivo experiments. Taken together, these data show that downregulating LINC00319 in CD133+CD144+ TU177 cells may serve as a potential anticancer regimen by inhibiting the proliferation and invasion of cancer stem cells in LSCC.


Asunto(s)
Carcinoma de Células Escamosas/genética , Factor de Transcripción E2F1/genética , Regulación Neoplásica de la Expresión Génica , Proteína HMGB3/genética , Neoplasias Laríngeas/genética , Células Madre Neoplásicas/patología , ARN Largo no Codificante/metabolismo , Regulación hacia Arriba/genética , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Factor de Transcripción E2F1/metabolismo , Proteína HMGB3/metabolismo , Humanos , Neoplasias Laríngeas/patología , Ratones Endogámicos BALB C , Ratones Desnudos , Modelos Biológicos , Células Madre Neoplásicas/metabolismo , Pronóstico , ARN Largo no Codificante/genética
15.
BMC Pulm Med ; 21(1): 191, 2021 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-34092219

RESUMEN

BACKGROUND: Long noncoding RNAs contribute to various inflammatory diseases, including sepsis. We explore the role of small nucleolar RNA host gene 16 (SNHG16) in sepsis-mediated acute lung injury (ALI) and inflammation. METHODS: A sepsis-induced ALI rat model was constructed by the cecal ligation and perforation method. The profiles of SNHG16, miR-128-3p, and high-mobility group box 3 (HMGB3) were monitored by quantitative reverse transcription PCR and Western blot. The pathologic changes of lung tissues were evaluated by Hematoxylin-Eosin staining, immunohistochemistry, and dry and wet method. Meanwhile, the pro-inflammatory factors and proteins were determined by ELISA and Western blot. In contrast, a sepsis model in BEAS-2B was induced with lipopolysaccharide (LPS) to verify the effects of SNHG16/miR-128-3p/HMGB3 on lung epithelial cell viability and apoptosis. RESULTS: As a result, SNHG16 and HMGB3 were up-regulated, while miR-128-3p was down-regulated in sepsis-induced ALI both in vivo and in vitro. Inhibiting SNHG16 reduced the apoptosis and inflammation in the sepsis-induced ALI model. Overexpressing SNHG16 promoted LPS-mediated lung epithelial apoptosis and inhibited cell viability and inflammation, while miR-128-3p had the opposite effects. Mechanistically, SNHG16 targeted miR-128-3p and attenuated its expression, while miR-128-3p targeted the 3' untranslated region of HMGB3. CONCLUSIONS: Overall, down-regulating SNHG16 alleviated the sepsis-mediated ALI by regulating miR-128-3p/HMGB3.


Asunto(s)
Lesión Pulmonar Aguda/genética , Proteína HMGB3/genética , MicroARNs/genética , ARN Largo no Codificante/genética , Sepsis/genética , Animales , Apoptosis , Supervivencia Celular , Regulación hacia Abajo , Regulación de la Expresión Génica , Humanos , Lipopolisacáridos/farmacología , Masculino , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba
16.
Biosci Rep ; 41(5)2021 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-33960364

RESUMEN

BACKGROUND: Esophageal cancer (ESCA) is one of the most commonly diagnosed cancers in the world. Tumor immune microenvironment is closely related to tumor prognosis. The present study aimed at analyzing the competing endogenous RNA (ceRNA) network and tumor-infiltrating immune cells in ESCA. METHODS: The expression profiles of mRNAs, lncRNAs, and miRNAs were downloaded from the Cancer Genome Atlas database. A ceRNA network was established based on the differentially expressed RNAs by Cytoscape. CIBERSORT was applied to estimate the proportion of immune cells in ESCA. Prognosis-associated genes and immune cells were applied to establish prognostic models basing on Lasso and multivariate Cox analyses. The survival curves were constructed with Kaplan-Meier method. The predictive efficacy of the prognostic models was evaluated by the receiver operating characteristic (ROC) curves. RESULTS: The differentially expressed mRNAs, lncRNAs, and miRNAs were identified. We constructed the ceRNA network including 23 lncRNAs, 19 miRNAs, and 147 mRNAs. Five key molecules (HMGB3, HOXC8, HSPA1B, KLHL15, and RUNX3) were identified from the ceRNA network and five significant immune cells (plasma cells, T cells follicular helper, monocytes, dendritic cells activated, and neutrophils) were selected via CIBERSORT. The ROC curves based on key genes and significant immune cells all showed good sensitivity (AUC of 3-year survival: 0.739, AUC of 5-year survival: 0.899, AUC of 3-year survival: 0.824, AUC of 5-year survival: 0.876). There was certain correlation between five immune cells and five key molecules. CONCLUSION: The present study provides an effective bioinformatics basis for exploring the potential biomarkers of ESCA and predicting its prognosis.


Asunto(s)
Células Dendríticas/metabolismo , Neoplasias Esofágicas/genética , Neutrófilos/metabolismo , Linfocitos T/metabolismo , Transcriptoma , Microambiente Tumoral , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Neoplasias Esofágicas/inmunología , Neoplasias Esofágicas/patología , Proteína HMGB3/genética , Proteína HMGB3/metabolismo , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
17.
Int J Biochem Cell Biol ; 135: 105967, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33727043

RESUMEN

OBJECTIVE: Histone deacetylase 3 (HDAC3) has been reported to repress the expression of various genes by eliminating acetyl group from histone. The objective of this study was to discuss the effect of HDAC3/microRNA-130a-3p (miR-130a-3p)/high-mobility group box 3 (HMGB3) on immune escape of breast cancer. METHODS: HDAC3, miR-130a-3p and HMGB3 expression in breast cancer tissues and cells were tested, and the correlation between HDAC3, miR-130a-3p and HMGB3 was analyzed. CD8, CD69 and programmed cell death protein 1 (PD-1) expression was detected. MDA-MB-231 cells were treated with relative plasmid of HDAC3 or miR-130a-3p to test cell viability, migration, epithelial-mesenchymal transition (EMT) and apoptosis in MDA-MB-231 cells. The cytotoxicity of CD8+/CD69+/PD-1+T cells in MDA-MB-231 cells was tested, and CD8+/CD69+/PD-1+T cell proliferation and apoptosis before and after co-culture with MDA-MB-231 cells were detected. RESULTS: HDAC3 and HMGB3 expression were raised and miR-130a-3p expression was diminished in breast cancer tissues and cells. HDAC3 was negatively correlated with miR-130a-3p while miR-130a-3p was negatively correlated with HMGB3. Down-regulating HDAC3 or up-regulating miR-130a-3p restrained cell viability, migration, EMT and anti-CD8+/CD69+/PD-1+T cytotoxicity and facilitated apoptosis of breast cancer cells. HDAC3 regulated HMGB3 by mediating miR-130a-3p expression. Down-regulating miR-130a-3p reversed the role of HDAC3 reduction on breast cancer cells. HDAC3 regulated CD8+/CD69+/PD-1+T cell proliferation and apoptosis by mediating miR-130a-3p. CONCLUSION: This study provides evidence that HDAC3 increases HMGB3 expression to promote the immune escape of breast cancer cells via down-regulating miR-130a-3p.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/inmunología , Regulación Neoplásica de la Expresión Génica , Proteína HMGB3/metabolismo , Histona Desacetilasas/metabolismo , MicroARNs/genética , Escape del Tumor , Apoptosis , Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Movimiento Celular , Proliferación Celular , Transición Epitelial-Mesenquimal , Femenino , Proteína HMGB3/genética , Histona Desacetilasas/genética , Humanos , Pronóstico , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Exp Mol Pathol ; 120: 104635, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33773992

RESUMEN

OBJECTIVE: MicroRNA (miR)-93 has been proven to mediate the initiation and progression of colorectal carcinoma (CRC); however, the mechanisms by which miR-93 mediates CRC development need deeper elucidation. The present study is designed to investigate the association between miR-93 and high mobility group box 3 (HMGB3), as well as the functions of miR-93, in CRC. METHODS: miR-93 expression was quantified by RT-qPCR. CRC cells were transfected or cotransfected with miR-93 mimic, miR-93 inhibitor, pcDNA3.1-HMGB3 and sh-HMGB3, and then the proliferative, migratory and invasive capacities were detected in addition to the apoptotic rate. Western blotting assessed the expression levels of HMGB3, PI3K, p-PI3K, AKT and p-AKT. The interaction between miR-93 and HMGB3 was identified. RESULTS: In CRC tissues, miR-93 was downregulated and HMGB3 was upregulated. LOVO and SW480 cells transfected with miR-93 mimic exhibited reduced proliferation, invasion and migration as well as increased apoptosis. The ratios of p-PI3K/PI3K and p-AKT/AKT were declined after miR-93 mimic was introduced into the CRC cell lines. miR-93 negatively downregulated HMGB3, and introduction of pcDNA3,1-HMGB3 could counteract, in part, the inhibitory effects of miR-93 on the malignant properties of CRC cells as well as the ratios of p-PI3K/PI3K and p-AKT/AKT. CONCLUSION: miR-93 targeted HMGB3 to block the activation of the PI3K/AKT pathway and thus enhance CRC cell apoptosis.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Colorrectales/patología , Regulación Neoplásica de la Expresión Génica , Proteína HMGB3/metabolismo , MicroARNs/genética , Apoptosis , Biomarcadores de Tumor/genética , Movimiento Celular , Proliferación Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Femenino , Proteína HMGB3/genética , Humanos , Masculino , Persona de Mediana Edad , Invasividad Neoplásica , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Pronóstico , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Células Tumorales Cultivadas
19.
IUBMB Life ; 72(11): 2508-2524, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32918539

RESUMEN

Circular RNAs (circRNAs) are non-coding RNAs that have shown to regulate the progression of human diseases, including a variety of cancers. We aimed to investigate the function and the underlying working mechanism of circRNA matrix metallopeptidase 1 (circMMP1; hsa_circ_0024108) in glioma progression. Quantitative real-time polymerase chain reaction (qRT-PCR) was performed to examine the expression of circMMP1, microRNA-433-3p (written as "miR-433") and high mobility group box 3 (HMGB3). Nanoparticle tracking analysis (NTA) was used to analyze the relative concentration and size distribution of serum exosomes. Cell proliferation was analyzed via cell counting kit-8 (CCK8) assay and colony formation assay. Transwell migration and invasion assays were used to examine the metastasis ability of glioma cells. Cell apoptosis was assessed by flow cytometry. Dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay were used to verify the interactions of circMMP1/miR-433 and miR-433/HMGB3 messenger RNA (mRNA). Xenograft mouse model was built to clarify the role of circMMP1 in vivo. The results showed that high serum exosomal circMMP1 level might predict poor prognosis of glioma patients. CircMMP1 promoted the proliferation and motility and impeded the apoptosis of glioma cells. MiR-433 was a direct target of circMMP1, and circMMP1 silencing-mediated influences in glioma cells were partly alleviated by the knockdown of miR-433. MiR-433 directly interacted with HMGB3 mRNA, and HMGB3 overexpression partly counteracted the effects of miR-433 up-regulation in glioma cells. CircMMP1 enhanced HMGB3 level through sponging miR-433 in glioma cells. CircMMP1 silencing suppressed the progression of glioma in vivo. CircMMP1 promoted the progression of glioma through acting as a competitive endogenous RNA (ceRNA) of miR-433 to up-regulate HMGB3. CircMMP1 level in serum exosomes might be a potential marker for the diagnosis of glioma patients. Targeting circMMP1/miR-433/HMGB3 signaling might be a novel insight for glioma treatment.


Asunto(s)
Neoplasias Encefálicas/patología , Regulación Neoplásica de la Expresión Génica , Glioma/patología , Proteína HMGB3/metabolismo , Metaloproteinasa 1 de la Matriz/genética , MicroARNs/genética , ARN Circular/genética , Animales , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Proliferación Celular , Glioma/genética , Glioma/metabolismo , Proteína HMGB3/genética , Humanos , Técnicas In Vitro , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Pronóstico , Tasa de Supervivencia , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Pathol Res Pract ; 216(11): 153144, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32911346

RESUMEN

Non-small cell lung cancer (NSCLC) accounting for 85 % of all lung cancer was one of the main causes of death worldwide. In this study, we investigated the role of circRNA_102179 in NSCLC development. The levels of circRNA_102179 in NSCLC tissues and cell lines were determined by quantitative real-time PCR assay (qRT-PCR). CCK8 and colony formation assays were applied to explore the effect of circRNA_102179 on the growth of NSCLC cells in vitro. Transwell assay was utilized to analyze the impact of circRNA_102179 on the migration and invasion of NSCLC cells. Target prediction and luciferase reporter assay were used to identify the interacting miRNA of circRNA_102179. The interaction among circRNA_102179/ miR-330-5p/HMGB3 was further validated by colony formation and Transwell invasion assays. Finally, the mouse xenograft NSCLC model was used to explore the role of circRNA_102179 in the tumor growth of NSCLC cells in vivo. CircRNA_102179 was overexpressed in NSCLC tissues and cells compared with normal lung tissues and human bronchial epithelial cells (HBEs). The down-regulation of circRNA_102179 markedly reduced the proliferation, migration, and invasion of NSCLC cells. Moreover, down-expression of circRNA_102179 significantly increased the level of miR-330-5p/HMGB3 in NSCLC cells. Further functional experiments indicated that over-expression of miR-330-5p reversed the inhibitory effect of circRNA_102179 on NSCLC cells growth, migration, and invasion. Our results reveal that circRNA_102179 facilitates the proliferation, migration, and invasion of NSCLC cell via modulating miR-330-5p/ HMGB3 axis in NSCLC cells.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/patología , Proteína HMGB3/genética , Neoplasias Pulmonares/patología , MicroARNs/genética , ARN Circular/genética , Animales , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Movimiento Celular/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica/fisiología , Proteína HMGB3/metabolismo , Xenoinjertos , Humanos , Neoplasias Pulmonares/genética , Ratones , Invasividad Neoplásica/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...