Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 229
Filtrar
1.
Mol Cell ; 70(4): 730-744.e6, 2018 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-29706538

RESUMEN

Processes like cellular senescence are characterized by complex events giving rise to heterogeneous cell populations. However, the early molecular events driving this cascade remain elusive. We hypothesized that senescence entry is triggered by an early disruption of the cells' three-dimensional (3D) genome organization. To test this, we combined Hi-C, single-cell and population transcriptomics, imaging, and in silico modeling of three distinct cells types entering senescence. Genes involved in DNA conformation maintenance are suppressed upon senescence entry across all cell types. We show that nuclear depletion of the abundant HMGB2 protein occurs early on the path to senescence and coincides with the dramatic spatial clustering of CTCF. Knocking down HMGB2 suffices for senescence-induced CTCF clustering and for loop reshuffling, while ectopically expressing HMGB2 rescues these effects. Our data suggest that HMGB2-mediated genomic reorganization constitutes a primer for the ensuing senescent program.


Asunto(s)
Factor de Unión a CCCTC/metabolismo , Cromatina/metabolismo , Genoma Humano , Proteína HMGB2/metabolismo , Factor de Unión a CCCTC/genética , Proliferación Celular , Senescencia Celular , Cromatina/genética , Proteína HMGB2/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos
2.
Nat Immunol ; 14(2): 179-85, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23263557

RESUMEN

The innate immune system senses viral DNA that enters mammalian cells, or in aberrant situations self-DNA, and triggers type I interferon production. Here we present an integrative approach that combines quantitative proteomics, genomics and small molecule perturbations to identify genes involved in this pathway. We silenced 809 candidate genes, measured the response to dsDNA and connected resulting hits with the known signaling network. We identified ABCF1 as a critical protein that associates with dsDNA and the DNA-sensing components HMGB2 and IFI204. We also found that CDC37 regulates the stability of the signaling molecule TBK1 and that chemical inhibition of the CDC37-HSP90 interaction and several other pathway regulators potently modulates the innate immune response to DNA and retroviral infection.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/inmunología , ADN Viral/inmunología , Células Dendríticas/inmunología , Fibroblastos/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Inmunidad Innata , Transportadoras de Casetes de Unión a ATP/genética , Animales , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/inmunología , Chaperoninas/antagonistas & inhibidores , Chaperoninas/genética , Chaperoninas/inmunología , Citosol/efectos de los fármacos , Citosol/metabolismo , Citosol/virología , ADN Viral/genética , Células Dendríticas/efectos de los fármacos , Células Dendríticas/virología , Fibroblastos/efectos de los fármacos , Fibroblastos/virología , Regulación de la Expresión Génica/inmunología , Silenciador del Gen , VIH-1/fisiología , Proteína HMGB2/genética , Proteína HMGB2/inmunología , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP90 de Choque Térmico/genética , Proteínas HSP90 de Choque Térmico/inmunología , Humanos , Ratones , Ratones Transgénicos , Proteínas Nucleares/genética , Proteínas Nucleares/inmunología , Fosfoproteínas/genética , Fosfoproteínas/inmunología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Proteómica , ARN Interferente Pequeño/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Bibliotecas de Moléculas Pequeñas/farmacología , Vesiculovirus/fisiología
3.
Arterioscler Thromb Vasc Biol ; 44(6): e172-e195, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38572649

RESUMEN

BACKGROUND: Pulmonary hypertension (PH) is a progressive and life-threatening disease characterized by pulmonary vascular remodeling, which involves aberrant proliferation and apoptosis resistance of the pulmonary arterial smooth muscle cells (PASMCs), resembling the hallmark characteristics of cancer. In cancer, the HMGB2 (high-mobility group box 2) protein promotes the pro-proliferative/antiapoptotic phenotype. However, the function of HMGB2 in PH remains uninvestigated. METHODS: Smooth muscle cell (SMC)-specific HMGB2 knockout or HMGB2-OE (HMGB2 overexpression) mice and HMGB2 silenced rats were used to establish hypoxia+Su5416 (HySu)-induced PH mouse and monocrotaline-induced PH rat models, respectively. The effects of HMGB2 and its underlying mechanisms were subsequently elucidated using RNA-sequencing and cellular and molecular biology analyses. Serum HMGB2 levels were measured in the controls and patients with pulmonary arterial (PA) hypertension. RESULTS: HMGB2 expression was markedly increased in the PAs of patients with PA hypertension and PH rodent models and was predominantly localized in PASMCs. SMC-specific HMGB2 deficiency or silencing attenuated PH development and pulmonary vascular remodeling in hypoxia+Su5416-induced mice and monocrotaline-treated rats. SMC-specific HMGB2 overexpression aggravated hypoxia+Su5416-induced PH. HMGB2 knockdown inhibited PASMC proliferation in vitro in response to PDGF-BB (platelet-derived growth factor-BB). In contrast, HMGB2 protein stimulation caused the hyperproliferation of PASMCs. In addition, HMGB2 promoted PASMC proliferation and the development of PH by RAGE (receptor for advanced glycation end products)/FAK (focal adhesion kinase)-mediated Hippo/YAP (yes-associated protein) signaling suppression. Serum HMGB2 levels were significantly increased in patients with PA hypertension, and they correlated with disease severity, predicting worse survival. CONCLUSIONS: Our findings indicate that targeting HMGB2 might be a novel therapeutic strategy for treating PH. Serum HMGB2 levels could serve as a novel biomarker for diagnosing PA hypertension and determining its prognosis.


Asunto(s)
Modelos Animales de Enfermedad , Proteína HMGB2 , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular , Miocitos del Músculo Liso , Arteria Pulmonar , Remodelación Vascular , Animales , Proteína HMGB2/genética , Proteína HMGB2/metabolismo , Humanos , Masculino , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Arteria Pulmonar/metabolismo , Arteria Pulmonar/fisiopatología , Arteria Pulmonar/patología , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Músculo Liso Vascular/fisiopatología , Ratas , Ratones , Proliferación Celular , Índice de Severidad de la Enfermedad , Transducción de Señal , Hipertensión Arterial Pulmonar/metabolismo , Hipertensión Arterial Pulmonar/fisiopatología , Ratas Sprague-Dawley , Femenino , Células Cultivadas , Persona de Mediana Edad , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/fisiopatología
4.
J Proteome Res ; 23(3): 1075-1087, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38376246

RESUMEN

Severe acute pancreatitis (SAP) is a highly fatal abdominal emergency, and its association with protein arginine methyltransferase 7 (PRMT7), the sole known type III enzyme responsible for the monomethylation of arginine residue, remains unexplored. In this study, we observe an increase in the PRMT7 levels in the pancreas of SAP mice and Cerulein-LPS-stimulated AR42J cells. Overexpression of Prmt7 exacerbated pancreatic damage in SAP, while the inhibition of PRMT7 improved SAP-induced pancreatic damage. Furthermore, PRMT7 overexpression promoted inflammation, oxidative stress, and ferroptosis during SAP. Mechanically, PRMT7 catalyzed monomethylation at histone H4 arginine 3 (H4R3me1) at the promoter region of high mobility group proteins 2 (HMGB2), thereby enhancing its transcriptional activity. Subsequently, HMGB2 facilitated Acyl CoA synthase long-chain family member 1 (ACSL1) transcription by binding to its promoter region, resulting in the activation of ferroptosis. Inhibition of PRMT7 effectively alleviated ferroptosis in Cerulein-LPS-induced AR42J cells by suppressing the HMGB2-ACSL1 pathway. Overall, our study reveals that PRMT7 plays a crucial role in promoting SAP through its regulation of the HMGB2-ACSL1 pathway to accelerate ferroptosis.


Asunto(s)
Ferroptosis , Pancreatitis , Animales , Ratones , Enfermedad Aguda , Arginina , Ceruletida , Ferroptosis/genética , Proteína HMGB2 , Lipopolisacáridos , Pancreatitis/inducido químicamente , Pancreatitis/genética , Proteína-Arginina N-Metiltransferasas/genética , Factores de Transcripción , Activación Transcripcional
5.
Ren Fail ; 46(1): 2338931, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38622929

RESUMEN

BACKGROUND: IgA nephropathy (IgAN) is the most common primary glomerulonephritis worldwide. Proliferation-inducing ligand (APRIL) was identified as an important cause of glycosylation deficiency of IgA1 (Gd-IgA1), which can 'trigger' IgAN. Our previous study indicated that high migration group protein B2 (HMGB2) in peripheral blood mononuclear cells from patients with IgAN was associated with disease severity, but the underlying mechanism remains unclear. MATERIALS AND METHODS: The location of HMGB2 was identified by immunofluorescence. qRT-PCR and Western blotting were used to measure HMGB2, HMGA1, and APRIL expression. Gd-IgA1 levels were detected by enzyme-linked immunosorbent assay (ELISA). In addition, we used DNA pull-down, protein profiling, and transcription factor prediction software to identify proteins bound to the promoter region of the APRIL gene. RNA interference and coimmunoprecipitation (Co-IP) were used to verify the relationships among HMGB2, high mobility group AT-hook protein 1 (HMGA1), and APRIL. RESULTS: HMGB2 expression was greater in IgAN patients than in HCs and was positively associated with APRIL expression in B cells. DNA pull-down and protein profiling revealed that HMGB2 and HMGA1 bound to the promoter region of the APRIL gene. The expression levels of HMGA1, APRIL, and Gd-IgA1 were downregulated after HMGB2 knockdown. Co-IP indicated that HMGB2 binds to HMGA1. The Gd-IgA1 concentration in the supernatant was reduced after HMGA1 knockdown. HMGA1 binding sites were predicted in the promoter region of the APRIL gene. CONCLUSION: HMGB2 expression is greater in IgAN patients than in healthy controls; it promotes APRIL expression by interacting with HMGA1, thereby inducing Gd-IgA1 overexpression and leading to IgAN.


Asunto(s)
Glomerulonefritis por IGA , Humanos , ADN/metabolismo , Glicosilación , Proteína HMGA1a/metabolismo , Proteína HMGB2/genética , Proteína HMGB2/metabolismo , Inmunoglobulina A , Leucocitos Mononucleares/metabolismo , Factores de Transcripción/metabolismo , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral
6.
Int J Mol Sci ; 25(6)2024 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-38542079

RESUMEN

Human high-mobility group-B (HMGB) proteins regulate gene expression in prostate cancer (PCa), a leading cause of oncological death in men. Their role in aggressive PCa cancers, which do not respond to hormonal treatment, was analyzed. The effects of HMGB1 and HMGB2 silencing upon the expression of genes previously related to PCa were studied in the PCa cell line PC-3 (selected as a small cell neuroendocrine carcinoma, SCNC, PCa model not responding to hormonal treatment). A total of 72% of genes analyzed, using pre-designed primer panels, were affected. HMGB1 behaved mostly as a repressor, but HMGB2 as an activator. Changes in SERPINE1, CDK1, ZWINT, and FN1 expression were validated using qRT-PCR after HMGB1 silencing or overexpression in PC-3 and LNCaP (selected as an adenocarcinoma model of PCa responding to hormonal treatment) cell lines. Similarly, the regulatory role of HMGB2 upon SERPINE1, ZWINT, FN1, IGFPB3, and TYMS expression was validated, finding differences between cell lines. The correlation between the expression of HMGB1, HMGB2, and their targets was analyzed in PCa patient samples and also in PCa subgroups, classified as neuroendocrine positive or negative, in public databases. These results allow a better understanding of the role of HMGB proteins in PCa and contribute to find specific biomarkers for aggressive PCa.


Asunto(s)
Adenocarcinoma , Proteína HMGB1 , Neoplasias de la Próstata , Humanos , Masculino , Adenocarcinoma/patología , Línea Celular , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Proteína HMGB2/genética , Proteína HMGB2/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Factores de Transcripción
7.
Cardiology ; 148(3): 271-277, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36958298

RESUMEN

INTRODUCTION: The aim of this study was to investigate the relationship between the high mobility group box-2 (HMGB2) and valve calcification in senile degenerative heart valve disease (SDHVD). METHODS: According to the echocardiographic results, patients with calcified heart valves were used as the experimental group and patients without calcified heart valves were used as the control group; blood was drawn for testing, and serum levels of HMGB2 were measured by an enzyme-linked immunosorbent assay. Human heart valve interstitial cells (hVICs) cultured in vitro were randomly divided into two groups. The calcification group was cultured with a medium containing calcification induction solution and cells were induced on days 1, 3, and 5, and the control group was cultured with a standard medium. Expression of bone morphogenetic protein 4 (BMP-4) and HMGB2 in both groups was detected by Western blot. RT-PCR was performed to detect the expression of the HMGB2 gene during calcification. The hVICs were cultured in vitro for 4 days with different concentrations of exogenous HMGB2 (0.01 µg/mL, 0.1 µg/mL, 1 µg/mL, 2 µg/mL), while the control group was cultured with a standard medium and the expression of BMP-4 and NF-κB P65 was detected by Western blot. RESULTS: The serum level of HMGB2 was 7.90 (5.92, 12.39) µg/L, higher than that of 7.06 (5.06, 9.73) µg/L in the valve calcification group in elderly patients with degenerative valve disease (p = 0.005); the differences were statistically significant. In in vitro experiments, the cellular calcification protein BMP-4 and the HMGB2 protein were higher in the calcification group compared to the control group (p < 0.05). Exogenous stimulation of hVICs with HMGB2 was able to upregulate the expression of BMP-4 and NF-κB P65 (p < 0.05). CONCLUSIONS: HMGB2 is correlated with valvular calcification in senile degenerative heart valve disease. The HMGB2 protein may promote the process of SDHVD valve calcification by activating the NF-κB pathway and upregulating the expression of BMP-4.


Asunto(s)
Estenosis de la Válvula Aórtica , Calcinosis , Enfermedades de las Válvulas Cardíacas , Humanos , Anciano , Válvula Aórtica/metabolismo , FN-kappa B/metabolismo , Proteína HMGB2/metabolismo , Enfermedades de las Válvulas Cardíacas/metabolismo , Células Cultivadas
8.
J Biochem Mol Toxicol ; 37(5): e23306, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36935520

RESUMEN

Osteoarthritis (OA) is the most common age-related joint disease characterized by chronic inflammation, progressive articular cartilage destruction, and subchondral sclerosis. Accumulating evidence suggests that circular RNAs (circRNAs) play key roles in OA, but the function of circSLTM in OA remains greatly unknown. Therefore, this study focused on interleukin-1ß (IL-1ß)-treated primary human chondrocytes as well as a rat model to investigate the expression pattern and functional role of circSLTM in OA in vitro and in vivo. CircSLTM and high mobility group protein B2 (HMGB2) were upregulated in IL-1ß-induced chondrocytes, whereas miR-421 was downregulated. Knockdown of circSLTM or overexpression of miR-421 ameliorated IL-1ß-induced chondrocyte apoptosis and inflammation. The regulatory relationship between circSLTM and miR-421, as well as that between miR-421 and HMGB2, was predicted by bioinformatics and then verified by the RNA immunoprecipitation experiment and dual-luciferase reporter gene assay. Furthermore, silencing of circSLTM increased cartilage destruction and decreased cartilage tissue apoptosis rate and inflammation in a rat model of OA. Taken together, our findings demonstrate the fundamental role of circSLTM in OA progression and provide a potential molecular target for OA therapy.


Asunto(s)
MicroARNs , Osteoartritis , Humanos , Ratas , Animales , Condrocitos/metabolismo , ARN Circular/genética , ARN Circular/metabolismo , Proteína HMGB2/genética , Proteína HMGB2/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Inflamación/metabolismo , Osteoartritis/metabolismo , Factores de Transcripción/metabolismo , Interleucina-1beta/metabolismo , Apoptosis
9.
Mediators Inflamm ; 2023: 2157355, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38148870

RESUMEN

Background: Ferroptosis is a new form of cell death, which is closely related to the occurrence of many diseases. Our work focused on the mechanism by which HMGB2 regulate ferroptosis and inflammation in abdominal aortic aneurysm (AAA). Methods: Reverse transcription-quantitative polymerase chain reaction and western blot were utilized to assess HMGB2 levels. CCK-8 and flow cytometry assays were utilized to measure cell viability and apoptosis. We detected reactive oxygen species generation, Fe2+ level, and ferroptosis-related protein levels in Ang-II-treated VSMCs, which were typical characteristics of ferroptosis. Finally, the mice model of AAA was established to verify the function of HMGB2 in vivo. Results: Increased HMGB2 level was observed in Ang-II-treated VSMCs and Ang-II-induced mice model. HMGB2 depletion accelerated viability and impeded apoptosis in Ang-II-irritatived VSMCs. Moreover, HMGB2 deficiency neutralized the increase of ROS in VSMCs caused by Ang-II. HMGB2 silencing considerably weakened Ang-II-caused VSMC ferroptosis, as revealed by the decrease of Fe2+ level and ACSL4 and COX2 levels and the increase in GPX4 and FTH1 levels. Furthermore, the mitigation effects of shHMGB2 on Ang-II-induced VSMC damage could be counteracted by erastin, a ferroptosis agonist. Mechanically, HMGB2 depletion inactivated the NF-κß signaling in Ang-II-treated VSMCs. Conclusions: Our work demonstrated that inhibition of HMGB2-regulated ferroptosis and inflammation to protect against AAA via NF-κß signaling, suggesting that HMGB2 may be a potent therapeutic agent for AAA.


Asunto(s)
Aneurisma de la Aorta Abdominal , Ferroptosis , Ratones , Animales , Proteína HMGB2 , Aneurisma de la Aorta Abdominal/metabolismo , Factores de Transcripción , Inflamación/complicaciones , Angiotensina II/farmacología
10.
Int J Mol Sci ; 24(9)2023 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-37176041

RESUMEN

High-Mobility Group (HMG) chromosomal proteins are the most numerous nuclear non-histone proteins. HMGB domain proteins are the most abundant and well-studied HMG proteins. They are involved in variety of biological processes. HMGB1 and HMGB2 were the first members of HMGB-family to be discovered and are found in all studied eukaryotes. Despite the high degree of homology, HMGB1 and HMGB2 proteins differ from each other both in structure and functions. In contrast to HMGB2, there is a large pool of works devoted to the HMGB1 protein whose structure-function properties have been described in detail in our previous review in 2020. In this review, we attempted to bring together diverse data about the structure and functions of the HMGB2 protein. The review also describes post-translational modifications of the HMGB2 protein and its role in the development of a number of diseases. Particular attention is paid to its interaction with various targets, including DNA and protein partners. The influence of the level of HMGB2 expression on various processes associated with cell differentiation and aging and its ability to mediate the differentiation of embryonic and adult stem cells are also discussed.


Asunto(s)
Proteína HMGB1 , Proteína HMGB2 , Proteína HMGB2/genética , Proteína HMGB2/metabolismo , Proteína HMGB1/metabolismo , Proteínas HMGB/metabolismo , Factores de Transcripción , ADN/metabolismo , Proteínas Nucleares , Proteínas del Grupo de Alta Movilidad
11.
Int J Mol Sci ; 24(4)2023 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-36834988

RESUMEN

Non-histone nuclear proteins HMGB1 and HMGB2 (High Mobility Group) are involved in many biological processes, such as replication, transcription, and repair. The HMGB1 and HMGB2 proteins consist of a short N-terminal region, two DNA-binding domains, A and B, and a C-terminal sequence of glutamic and aspartic acids. In this work, the structural organization of calf thymus HMGB1 and HMGB2 proteins and their complexes with DNA were studied using UV circular dichroism (CD) spectroscopy. Post-translational modifications (PTM) of HMGB1 and HMGB2 proteins were determined with MALDI mass spectrometry. We have shown that despite the similar primary structures of the HMGB1 and HMGB2 proteins, their post-translational modifications (PTMs) demonstrate quite different patterns. The HMGB1 PTMs are located predominantly in the DNA-binding A-domain and linker region connecting the A and B domains. On the contrary, HMGB2 PTMs are found mostly in the B-domain and within the linker region. It was also shown that, despite the high degree of homology between HMGB1 and HMGB2, the secondary structure of these proteins is also slightly different. We believe that the revealed structural properties might determine the difference in the functioning of the HMGB1 and HMGB2 as well as their protein partners.


Asunto(s)
Proteína HMGB1 , Proteína HMGB2 , ADN/química , ADN/metabolismo , Proteínas del Grupo de Alta Movilidad , Proteína HMGB1/química , Proteína HMGB1/metabolismo , Proteína HMGB2/química , Proteína HMGB2/metabolismo , Factores de Transcripción , Unión Proteica , Animales , Bovinos
12.
Int J Mol Sci ; 24(8)2023 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-37108449

RESUMEN

Transposons are parasitic genetic elements that frequently hijack vital cellular processes of their host. HMGXB4 is a known Wnt signaling-regulating HMG-box protein, previously identified as a host-encoded factor of Sleeping Beauty (SB) transposition. Here, we show that HMGXB4 is predominantly maternally expressed, and marks both germinal progenitor and somatic stem cells. SB piggybacks HMGXB4 to activate transposase expression and target transposition to germinal stem cells, thereby potentiating heritable transposon insertions. The HMGXB4 promoter is located within an active chromatin domain, offering multiple looping possibilities with neighboring genomic regions. HMGXB4 is activated by ERK2/MAPK1, ELK1 transcription factors, coordinating pluripotency and self-renewal pathways, but suppressed by the KRAB-ZNF/TRIM28 epigenetic repression machinery, also known to regulate transposable elements. At the post-translational level, SUMOylation regulates HMGXB4, which modulates binding affinity to its protein interaction partners and controls its transcriptional activator function via nucleolar compartmentalization. When expressed, HMGXB4 can participate in nuclear-remodeling protein complexes and transactivate target gene expression in vertebrates. Our study highlights HMGXB4 as an evolutionarily conserved host-encoded factor that assists Tc1/Mariner transposons to target the germline, which was necessary for their fixation and may explain their abundance in vertebrate genomes.


Asunto(s)
Cromosomas , Elementos Transponibles de ADN , Animales , Elementos Transponibles de ADN/genética , Células Madre , Proteína HMGB2/metabolismo
13.
Mol Cancer ; 21(1): 110, 2022 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-35525959

RESUMEN

BACKGROUND: Previous studies have confirmed the oncogenic role of HMGB2 in various cancers, but the biological functions of HMGB2-derived circRNAs remain unknown. Thus, we intended to investigate the potential role of HMGB2-derived circRNAs in lung adenocarcinomas (LUAD) and squamous cell carcinomas (LUSC). METHODS: The expression profiles of HMGB2-derived circRNAs in LUAD and LUSC tissues and matched normal tissues were assessed using qRT-PCR. The role of circHMGB2 in the progression of the LUAD and LUSC was determined in vitro by Transwell, CCK-8, flow cytometry and immunohistochemistry assays, as well as in vivo in an immunocompetent mouse model and a humanized mouse model. In addition, in vivo circRNA precipitation assays, luciferase reporter assays and RNA pulldown assays were performed to explore the underlying mechanism by which circHMGB2 promotes anti-PD-1 resistance in the LUAD and LUSC. RESULTS: The expression of circHMGB2 (hsa_circ_0071452) was significantly upregulated in NSCLC tissues, and survival analysis identified circHMGB2 as an independent indicator of poor prognosis in the LUAD and LUSC patients. We found that circHMGB2 exerted a mild effect on the proliferation of the LUAD and LUSC cells, but circHMGB2 substantially reshaped the tumor microenvironment by contributing to the exhaustion of antitumor immunity in an immunocompetent mouse model and a humanized mouse model. Mechanistically, circHMGB2 relieves the inhibition of downstream CARM1 by sponging miR-181a-5p, thus inactivating the type 1 interferon response in the LUAD and LUSC. Moreover, we found that the upregulation of circHMGB2 expression decreased the efficacy of anti-PD-1 therapy, and we revealed that the combination of the CARM1 inhibitor EZM2302 and an anti-PD-1 antibody exerted promising synergistic effects in a preclinical model. CONCLUSION: circHMGB2 overexpression promotes the LUAD and LUSC progression mainly by reshaping the tumor microenvironment and regulating anti-PD-1 resistance in the LUAD and LUSC patients. This study provides a new strategy for the LUAD and LUSC treatment.


Asunto(s)
Adenocarcinoma del Pulmón , Carcinoma de Células Escamosas , Neoplasias Pulmonares , MicroARNs , Proteína-Arginina N-Metiltransferasas , Adenocarcinoma del Pulmón/tratamiento farmacológico , Adenocarcinoma del Pulmón/genética , Animales , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/genética , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Proteína HMGB2/genética , Humanos , Terapia de Inmunosupresión , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Ratones , MicroARNs/genética , Proteína-Arginina N-Metiltransferasas/genética , ARN Circular/genética , Microambiente Tumoral
14.
J Gene Med ; 24(6): e3308, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-33395735

RESUMEN

BACKGROUND: The importance of long non-coding RNAs (lncRNAs) in the regulation of tumorigenesis has been gradually recognized. The roles of lncRNA cancer susceptibility candidate 15 (CASC15) in cancers have been validated by several independent groups; however, its role in colorectal cancer (CRC) remains to be explored. METHODS: Levels of CASC15 in CRC cells and normal cells were measured using a quantitative real-time polymerase chain reaction method. In vitro functional assays were performed to detect the effects of CASC15 on cell proliferation, invasion and apoptosis. Bioinformatic analyses and luciferase activity assays were conducted to investigate the targets for CASC15. Animal experiments were conducted to analyze the effect of CASC15 on tumor growth in vivo. RESULTS: The CASC15 level is revealed to be significantly elevated in CRC cells compared to that in normal cells. In vitro assays revealed that CASC15 overexpression stimulates cell growth and invasion, whereas its down-expression has opposite effects. Furthermore, CASC15 can bind with microRNA-582-5p (miR-582-5p) to modulate high mobility group box 2 (HMGB2) expression. We also showed that silencing of CASC15 inhibits tumor growth. CONCLUSIONS: In summary, CASC15 overexpression could promote CRC carcinogenesis, indicating that knockdown of CASC15 might be a possible therapeutic measure to hinder carcinogenesis. The results of the present study could help us to understand the mechanisms behind CRC progression.


Asunto(s)
Neoplasias Colorrectales , MicroARNs , ARN Largo no Codificante , Animales , Carcinogénesis/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Neoplasias Colorrectales/patología , Regulación Neoplásica de la Expresión Génica , Proteína HMGB2/genética , Proteína HMGB2/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , ARN Largo no Codificante/metabolismo , Factores de Transcripción/genética
15.
Histochem Cell Biol ; 157(3): 359-369, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35024954

RESUMEN

High-mobility group box 2 (HMGB2) is a chromatin-associated protein that is an important regulator of gene transcription, recombination, and repair processes. The functional importance of HMGB2 has been reported in various organs, including the testis, heart, and cartilage. However, its role in the ovary is largely unknown. In this study, ovary tissues from wild-type (WT) and HMGB2-knock-out (KO) mice were examined by histopathological staining and immunohistochemistry. The ovary size and weight were significantly lower in HMGB2-KO mice than in age-matched WT littermates. Histopathological analysis revealed ovarian atrophy and progressive fibrosis in 10-month-old HMGB2-KO mouse ovaries. Compared to age-matched WT mice, the numbers of oocytes and developing follicles were significantly decreased at 2 months of age and were completely depleted at 10 months of age in HMGB2-KO mice. Immunohistochemistry revealed the expression of HMGB2 in the granulosa cells of developing follicles, oocytes, some corpora lutea, and stromal cells. Importantly, HMGB2-positive cells were co-localized with estrogen receptor beta (ERß), but not ERα. Estrogen response element-binding activity was demonstrated by southwestern histochemistry, and it was decreased in HMGB2-KO mouse ovaries. Cell proliferation activity was also decreased in HMGB2-KO mouse ovaries in parallel with the decreased folliculogenesis. These results indicated that the depletion of HMGB2 induced ovarian atrophy that was characterized by a decreased ovarian size and weight, progressive fibrosis, as well as decreased oocytes and folliculogenesis. In conclusion, we demonstrated the crucial role of HMGB2 in mouse ovarian folliculogenesis through ERß expression.


Asunto(s)
Receptor beta de Estrógeno , Proteína HMGB2 , Animales , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/análisis , Receptor beta de Estrógeno/genética , Receptor beta de Estrógeno/metabolismo , Femenino , Células de la Granulosa , Proteína HMGB2/análisis , Proteína HMGB2/genética , Proteína HMGB2/metabolismo , Ratones , Ratones Noqueados , Ovario/metabolismo
16.
Fish Shellfish Immunol ; 127: 855-865, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35850457

RESUMEN

High mobility group box 1 (HMGB1) and HMGB2 have been demonstrated to be key regulators not only in DNA recombination, replication, gene transcription, but also in host inflammation and immune responses. In the present study, orthologs of HMGB1 and HMGB2 named Lc-HMGB1 and Lc-HMGB2 were characterized in large yellow croaker (Larimichthys crocea). The ORFs of Lc-HMGB1 and Lc-HMGB2 are 621 bp and 648 bp, encoding proteins of 206 aa and 215 aa, with the putative Lc-HMGB1 and Lc-HMGB2 proteins both contain two HMG domains, respectively. The genome organizations of Lc-HMGB1 and Lc-HMGB2 are both composed of four exons and three introns, which are conserved in vertebrates. Lc-HMGB1 and Lc-HMGB2 were identified as cell nucleus localized proteins, and were ubiquitously distributed in the examined organs/tissues. Additionally, Lc-HMGB1 was significantly up-regulated under LPS and PGN stimulation, whereas the stimulation of poly I:C, LPS, PGN, and Pseudomonas plecoglossicida infection could significantly induce Lc-HMGB2 expression in vivo. Notably, both Lc-HMGB1 and Lc-HMGB2 overexpression could significantly up-regulated the expression of diverse immune-related genes, including IFN1, IRF3, ISG15, ISG56, RSAD2, g-type lysozyme, and TNF-α. Moreover, overexpression of Lc-HMGB1 could also induce the expression of IRF7 and Mx. These results collectively indicate that Lc-HMGB1 and Lc-HMGB2 play important roles in host immune responses against pathogen infection.


Asunto(s)
Proteína HMGB1 , Perciformes , Animales , Clonación Molecular , Proteínas de Peces , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Proteína HMGB2/genética , Lipopolisacáridos/metabolismo , Lipopolisacáridos/farmacología , Filogenia
17.
Int J Clin Oncol ; 27(12): 1891-1903, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36269529

RESUMEN

BACKGROUND: Multiple studies have revealed that long non-coding RNA (lncRNA) NR2F2-AS1 plays a role in affecting cancer cell proliferation and metastasis. Here, both in vitro and in vivo experiments were performed for investigating the function and mechanism of NR2F2-AS1 in human osteosarcoma (OS). METHODS: The NR2F2-AS1 level in human OS tissues and adjacent non-tumor tissues was examined by quantitative reverse transcription-polymerase chain reaction (qRT-PCR). The NR2F2-AS1 overexpression model was constructed in OS cells, then cell proliferation, invasion, and apoptosis were monitored. The OS xenograft model was established in nude mice using NR2F2-AS1-overexpressed OS cells. The downstream target genes of NR2F2-AS1 were predicted. qRT-PCR and Western blot were implemented to validate the profiles of miR-425-5p and HMGB2. The targeting link between NR2F2-AS1 and miR-425-5p, miR-425-5p and HMGB2 was further probed by dual-luciferase reporter experiment. RESULTS: In comparison to adjacent non-tumor tissues, OS tissues showed upregulated NR2F2-AS1 expression. Higher NR2F2-AS1 level was predominantly correlated with worse clinical stages. In vivo and in vitro tests corroborated that NR2F2-AS1 overexpression spurred OS cell proliferation, growth, invasion, and choked apoptosis. Mechanistically, NR2F2-AS1 hampered miR-425-5p expression as its competitive endogenous RNA (ceRNA). Thus, NR2F2-AS1 facilitated the HMGB2 expression. However, miR-425-5p inhibited HMGB2 expression by targeting the latter. CONCLUSION: NR2F2-AS1 expedited the evolution of OS by elevating HMGB2 levels through sponging miR-425-5p. The NR2F2-AS1/miR-425-5p/HMGB2 regulatory axis is a promising target in treating human OS.


Asunto(s)
Neoplasias Óseas , Proteína HMGB2 , MicroARNs , Osteosarcoma , ARN Largo no Codificante , Animales , Humanos , Ratones , Neoplasias Óseas/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Factor de Transcripción COUP II/genética , Factor de Transcripción COUP II/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Proteína HMGB2/genética , Proteína HMGB2/metabolismo , Ratones Desnudos , MicroARNs/genética , MicroARNs/metabolismo , Osteosarcoma/genética , Osteosarcoma/patología , ARN Largo no Codificante/genética , Factores de Transcripción/genética
18.
BMC Pulm Med ; 22(1): 310, 2022 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-35962344

RESUMEN

BACKGROUND: High mobility group protein B2 (HMGB2) is a multifunctional protein that plays various roles in different cellular compartments. Moreover, HMGB2 serves as a potential prognostic biomarker and therapeutic target for lung adenocarcinoma (LUAD). METHODS: In this study, the expression pattern, prognostic implication, and potential role of HMGB2 in LUAD were evaluated using the integrated bioinformatics analyses based on public available mRNA expression profiles from The Cancer Genome Atlas and Gene Expression Omnibus databases, both at the single-cell level and the tissue level. Further study in the patient-derived samples was conducted to explore the correlation between HMGB2 protein expression levels with tissue specificity, (tumor size-lymph node-metastasis) TNM stage, pathological grade, Ki-67 status, and overall survival. In vitro experiments, such as CCK-8, colony-formation and Transwell assay, were performed with human LUAD cell line A549 to investigate the role of HMGB2 in LUAD progression. Furthermore, xenograft tumor model was generated with A549 in nude mice. RESULTS: The results showed that the HMGB2 expression was higher in the LUAD samples than in the adjacent normal tissues and was correlated with high degree of malignancy in different public data in this study. Besides, over-expression of HMGB2 promoted A549 cells proliferation and migration while knocking down of HMGB2 suppressed the tumor promoting effect. CONCLUSIONS: Our study indicated that HMGB2 was remarkably highly expressed in LUAD tissues, suggesting that it is a promising diagnostic and therapeutic marker for LUAD in the future.


Asunto(s)
Adenocarcinoma del Pulmón , Adenocarcinoma , Neoplasias Pulmonares , Adenocarcinoma/patología , Adenocarcinoma del Pulmón/patología , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Proteína HMGB2/genética , Humanos , Neoplasias Pulmonares/patología , Ratones , Ratones Desnudos , Pronóstico , Factores de Transcripción
19.
Mediators Inflamm ; 2022: 1376959, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35966335

RESUMEN

Cerebral infarction is a common disease with a higher disability and fatality rates. The incidence rates of cerebral infarction or cerebral ischemic stroke gradually increase with aging and cerebrovascular disease progression. This study is aimed at evaluating the effects of HMGA2 on cerebral infarction-induced brain tissue damage and its underlying mechanisms. Adult Sprague Dawley rats were pretreated with sh-HMGA2 before cerebral infarction operation. The effect of HMGA2 on the arrangement, distribution, and morphological structure of neurons and the cell apoptosis ratio in brain tissue were detected via hematoxylin and eosin staining, brain-water content, TTC staining, and TUNEL staining. The results from ELISA assay, qPCR, and western blot indicated that downregulation of HMGA2 mitigated inflammatory stress via regulating the expression of TLR4/NF-κB. In addition, results showed that suppressed HMGA2 attenuated the neurological dysfunction of brain injury rats and markedly reduced infarct volume. HMGA2 might be able to alleviate the damage associated with cerebral infarction-induced inflammatory response and cell apoptosis. Moreover, downregulation of HMGA2 had a protective effect on the brain damage derived from cerebral infarction by mediating the TLR4/NF-κB pathway. In conclusion, the current study demonstrated that downregulation of HMGB2 decreased the infarct size, inflammatory responses, and apoptosis in cerebral injury and further had neuroprotective effects against cerebral infarction-induced brain damage. Finally, these results indicated that the downregulation of the TLR4/NF-κB pathway after ischemia by HMGB2 inhibition is a potential mechanism of the neuroprotective effect of cerebral injury.


Asunto(s)
Lesiones Encefálicas , Isquemia Encefálica , Proteína HMGA2/metabolismo , Fármacos Neuroprotectores , Accidente Cerebrovascular , Animales , Isquemia Encefálica/metabolismo , Infarto Cerebral , Proteína HMGB2/metabolismo , FN-kappa B/metabolismo , Fármacos Neuroprotectores/farmacología , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Receptor Toll-Like 4/metabolismo
20.
Biochem Genet ; 60(6): 2489-2502, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35554782

RESUMEN

HMGB2, a member of the high-mobility group (HMG) proteins, was identified as a male-biased gene and plays a crucial role in the germ cells differentiation of mammals. However, its role in spermatogenesis of turtle is still poorly understood. Here, we cloned the Pelodiscus sinensis HMGB2 and analyzed its expression profile in different tissues and in testis at different developmental ages. P. sinensis HMGB2 mRNA was highly expressed in the testis of 3-year-old turtles (P < 0.01), but was hardly detected in ovaries and other somatic tissues. The results of chemical in situ hybridization (CISH) showed that HMGB2 mRNA was specifically expressed in germ cells, where it was mainly distributed in round spermatids and sperm, but not detected in somatic cells, spermatogonia, primary spermatocytes, or secondary spermatocyte. The relative expression of HMGB2 also responded to seasonal changes in testis development in P. sinensis. In different seasons of the year, the relative expression of HMGB2 transcripts in the testis of 1 year and 2 year olds showed an overall upward trend, whereas, in the testis of 3 year old, it peaked in July and then declined in October. Moreover, in April and July, with an increase in ages, the expression of HMGB2 transcripts showed an upward trend. However, in January and October, there was a decline in expression in testis in 3-year-old turtles. These results showed that HMGB2 is closely related to spermatogenesis in P. sinensis.


Asunto(s)
Tortugas , Animales , Masculino , Tortugas/genética , Tortugas/metabolismo , Proteína HMGB2/genética , Proteína HMGB2/metabolismo , Estaciones del Año , Semen , Espermatogénesis/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , China , Mamíferos/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda