Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 250
Filtrar
1.
Dis Markers ; 2022: 4487435, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35280439

RESUMO

Background: Receptor for advanced glycation end products (RAGE) is implicated in tumor biology. Released high mobility group box protein 1 (HMGB1) ligand binding to RAGE receptor in tumor cells promotes tumor progression. The mechanisms of HMGB1-RAGE signaling in M2 macrophages involved in lymphangiogenesis in laryngeal carcinoma remain poorly understood. Here, we assessed the effect of HMGB1-RAGE signaling on M2 macrophages in lymphangiogenesis. Methods: HMGB1, CD163, and D2-40 in laryngeal squamous cell carcinoma (LSCC, n = 123), laryngeal precursor lesions (LPLs, n = 102), and vocal polyp (VP, n = 55) were analyzed by immunohistochemistry. THP-1 cell-expressed RAGE gene was knocked down and then polarized to M0 macrophages and M2 macrophages. IL-23, TNF-α, TGF-ß, and IL-10 were measured by ELISA; IL-1ß, IL-12, IL-10, and CCL-13 were evaluated by RT-qPCR, and CD206, CD163, and RAGE were evaluated by western blot to evaluate whether classical M2 macrophages were obtained. Conditioned media from RAGE+/- M0 macrophages and RAGE+/- M2 macrophages incubated in the presence or absence of HMGB1, anti-Toll-like receptor (TLR)2, anti-TLR4 antibodies, and anti-VEGF-C antibodies were collected separately for human dermal lymphatic endothelial cells (HDLEC) for proliferation, migration, lymphangiogenesis assay, and VEGF-C concentration analysis. Results: HMGB1 and M2 macrophage densities were increased in LSCC (P < 0.01). HMGB1 and M2 macrophage densities were significantly correlated with lymphatic vessel density (LVD) in LSCC (P < 0.01). The HMGB1 overexpression and higher M2 macrophage density were involved in lymph node metastasis (P < 0.01) and poor prognosis (P < 0.05). In vitro, conditioned medium from HMGB1-stimulated RAGE+ M2 macrophages activated lymphangiogenesis by upregulating the VEGF compared to controls (P < 0.05). On the contrary, RAGE knockdown obviously decreased the corresponding effects of HMGB1-preconditioned M2 macrophages upon HDLEC (P < 0.05). HMGB1-TLR pathway does not significantly increase HDLEC proliferation, migration, and lymphangiogenesis on M2 macrophages. Conclusions: HMGB1 promotes lymphangiogenesis by activation of RAGE on M2 macrophages. Targeting RAGE may provide an effective therapeutic strategy against M2 macrophages in LSCC patients with lymph node metastasis.


Assuntos
Carcinoma de Células Escamosas/etiologia , Proteína HMGB1/fisiologia , Neoplasias Laríngeas/etiologia , Linfangiogênese , Macrófagos/fisiologia , Receptor para Produtos Finais de Glicação Avançada/fisiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
2.
J Mol Cell Cardiol ; 164: 69-82, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34838588

RESUMO

The global propagation of SARS-CoV-2 leads to an unprecedented public health emergency. Despite that the lungs are the primary organ targeted by COVID-19, systemic endothelial inflammation and dysfunction is observed particularly in patients with severe COVID-19, manifested by elevated endothelial injury markers, endotheliitis, and coagulopathy. Here, we review the clinical characteristics of COVID-19 associated endothelial dysfunction; and the likely pathological mechanisms underlying the disease including direct cell entry or indirect immune overreactions after SARS-CoV-2 infection. In addition, we discuss potential biomarkers that might indicate the disease severity, particularly related to the abnormal development of thrombosis that is a fatal vascular complication of severe COVID-19. Furthermore, we summarize clinical trials targeting the direct and indirect pathological pathways after SARS-CoV-2 infection to prevent or inhibit the virus induced endothelial disorders.


Assuntos
COVID-19/patologia , Endotélio Vascular/patologia , SARS-CoV-2 , Adolescente , Adulto , Idoso , Enzima de Conversão de Angiotensina 2/fisiologia , Animais , COVID-19/sangue , COVID-19/complicações , COVID-19/fisiopatologia , COVID-19/terapia , Ensaios Clínicos como Assunto , Células Endoteliais/patologia , Células Endoteliais/virologia , Endotélio Vascular/imunologia , Endotélio Vascular/fisiopatologia , Proteína HMGB1/fisiologia , Humanos , Macaca mulatta , Camundongos , Neuropilina-1/fisiologia , Estresse Oxidativo , Espécies Reativas de Oxigênio , Receptores Virais/fisiologia , Receptores Depuradores Classe B/fisiologia , Índice de Gravidade de Doença , Transdução de Sinais , Síndrome de Resposta Inflamatória Sistêmica/patologia , Síndrome de Resposta Inflamatória Sistêmica/fisiopatologia , Trombofilia/etiologia , Trombofilia/fisiopatologia , Fator A de Crescimento do Endotélio Vascular/fisiologia , Vasculite/etiologia , Vasculite/imunologia , Vasculite/fisiopatologia , Adulto Jovem
3.
Dis Markers ; 2021: 4933194, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34970357

RESUMO

Crosstalk between molecular regulators miR-126, hypoxia-inducible factor 1-alpha (HIF-1-α), and high-mobility group box-1 (HMGB1) contributes to the regulation of inflammation and angiogenesis in multiple physiological and pathophysiological settings. Here, we present evidence of an overriding role for miR-126 in the regulation of HMGB1 and its downstream proinflammatory effectors in endothelial cells subjected to hypoxia with concurrent acidosis (H/A). Methods. Primary mouse endothelial cells (PMEC) were exposed to hypoxia or H/A to simulate short or chronic low-flow ischemia, respectively. RT-qPCR quantified mRNA transcripts, and proteins were measured by western blot. ROS were quantified by fluorogenic ELISA and luciferase reporter assays employed to confirm an active miR-126 target in the HMGB1 3'UTR. Results. Enhanced expression of miR-126 in PMECs cultured under neutral hypoxia was suppressed under H/A, whereas the HMGB1 expression increased sequentially under both conditions. Enhanced expression of HMGB1 and downstream inflammation markers was blocked by the premiR-126 overexpression and optimized by antagomiR. Compared with neutral hypoxia, H/A suppressed the HIF-1α expression independently of miR-126. The results show that HMGB1 and downstream effectors are optimally induced by H/A relative to neutral hypoxia via crosstalk between hypoxia signaling, miR-126, and HIF-1α, whereas B-cell lymphoma 2(Bcl2), a HIF-1α, and miR-126 regulated gene expressed optimally under neutral hypoxia. Conclusion. Inflammatory responses of ECs to H/A are dynamically regulated by the combined actions of hypoxia, miR-126, and HIF-1α on the master regulator HMGB1. The findings may be relevant to vascular diseases including atherosclerotic occlusion and interiors of plaque where coexisting hypoxia and acidosis promote inflammation as a defining etiology.


Assuntos
Hipóxia Celular/fisiologia , Células Endoteliais/metabolismo , Proteína HMGB1/fisiologia , Inflamação/etiologia , MicroRNAs/fisiologia , Acidose , Animais , Células Cultivadas , Camundongos
4.
Front Immunol ; 12: 774807, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34925345

RESUMO

Radiation-induced lung injury (RILI) is a form of radiation damage to normal lung tissue caused by radiotherapy (RT) for thoracic cancers, which is most commonly comprised of radiation pneumonitis (RP) and radiation pulmonary fibrosis (RPF). Moreover, with the widespread utilization of immunotherapies such as immune checkpoint inhibitors as first- and second-line treatments for various cancers, the incidence of immunotherapy-related lung injury (IRLI), a severe immune-related adverse event (irAE), has rapidly increased. To date, we know relatively little about the underlying mechanisms and signaling pathways of these complications. A better understanding of the signaling pathways may facilitate the prevention of lung injury and exploration of potential therapeutic targets. Therefore, this review provides an overview of the signaling pathways of RILI and IRLI and focuses on their crosstalk in diverse signaling pathways as well as on possible mechanisms of adverse events resulting from combined radiotherapy and immunotherapy. Furthermore, this review proposes potential therapeutic targets and avenues of further research based on signaling pathways. Many new studies on pyroptosis have renewed appreciation for the value and importance of pyroptosis in lung injury. Therefore, the authors posit that pyroptosis may be the common downstream pathway of RILI and IRLI; discussion is also conducted regarding further perspectives on pyroptosis as a crucial signaling pathway in lung injury treatment.


Assuntos
Inibidores de Checkpoint Imunológico/efeitos adversos , Lesão Pulmonar/etiologia , Fibrose Pulmonar/etiologia , Pneumonite por Radiação/etiologia , Proteína HMGB1/fisiologia , Humanos , Fator 2 Relacionado a NF-E2/fisiologia , Piroptose , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/fisiologia
5.
Front Immunol ; 12: 705361, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34489957

RESUMO

Immunogenic cell death (ICD) is a form of regulated cell death (RCD) induced by various stresses and produces antitumor immunity via damage-associated molecular patterns (DAMPs) release or exposure, mainly including high mobility group box 1 (HMGB1), calreticulin (CRT), adenosine triphosphate (ATP), and heat shock proteins (HSPs). Emerging evidence has suggested that ionizing radiation (IR) can induce ICD, and the dose, type, and fractionation of irradiation influence the induction of ICD. At present, IR-induced ICD is mainly verified in vitro in mice and there is few clinical evidence about it. To boost the induction of ICD by IR, some strategies have shown synergy with IR to enhance antitumor immune response, such as hyperthermia, nanoparticles, and chemotherapy. In this review, we focus on the molecular mechanisms of ICD, ICD-promoting factors associated with irradiation, the clinical evidence of ICD, and immunogenic forms of cell death. Finally, we summarize various methods of improving ICD induced by IR.


Assuntos
Morte Celular Imunogênica/efeitos da radiação , Alarminas/fisiologia , Animais , Antígenos de Neoplasias/imunologia , Biomarcadores , Terapia Combinada , Citocinas/fisiologia , Relação Dose-Resposta à Radiação , Ferroptose/efeitos da radiação , Proteína HMGB1/fisiologia , Humanos , Hipertermia Induzida , Camundongos , Morfolinas/uso terapêutico , Necroptose/efeitos da radiação , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Neoplasias/radioterapia , Piperazinas/uso terapêutico , Pirróis/uso terapêutico , Tolerância a Radiação , Radiação Ionizante
6.
Front Immunol ; 12: 675731, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34234778

RESUMO

High mobility group box 1 (HMGB1) is a non-histone protein which is predominantly localised in the cell nucleus. However, stressed, dying, injured or dead cells can release this protein into the extracellular matrix passively. In addition, HMGB1 release was observed in cancer and immune cells where this process can be triggered by various endogenous as well as exogenous stimuli. Importantly, released HMGB1 acts as a so-called "danger signal" and could impact on the ability of cancer cells to escape host immune surveillance. However, the molecular mechanisms underlying the functional role of HMGB1 in determining the capability of human cancer cells to evade immune attack remain unclear. Here we report that the involvement of HMGB1 in anti-cancer immune evasion is determined by Toll-like receptor (TLR) 4, which recognises HMGB1 as a ligand. We found that HGMB1 induces TLR4-mediated production of transforming growth factor beta type 1 (TGF-ß), displaying autocrine/paracrine activities. TGF-ß induces production of the immunosuppressive protein galectin-9 in cancer cells. In TLR4-positive cancer cells, HMGB1 triggers the formation of an autocrine loop which induces galectin-9 expression. In malignant cells lacking TLR4, the same effect could be triggered by HMGB1 indirectly through TLR4-expressing myeloid cells present in the tumour microenvironment (e. g. tumour-associated macrophages).


Assuntos
Galectinas/biossíntese , Proteína HMGB1/fisiologia , Neoplasias/imunologia , Receptor 4 Toll-Like/fisiologia , Humanos , Tolerância Imunológica , Células THP-1 , Fator de Crescimento Transformador beta1/fisiologia
7.
Front Immunol ; 12: 675822, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34122437

RESUMO

We have reported that tumor-derived autophagosomes (DRibbles) were efficient carriers of tumor antigens and DRibbles antigens could be present by DRibbles-activated B cells to stimulate effect and naïve T cells in mice. However, the effect of DRibbles on human B cells remains unclear. Herein, we found that DRibbles can also efficiently induce proliferation and activation of human B cells and lead to the production of chemokines, cytokines and hematopoietic growth factors. We further demonstrated human B cells can effectively phagocytose DRibbles directly and cross-present DRibbles antigens to stimulate antigen-specific memory T cells. Furthermore, we found that membrane-bound high-mobility group B1 (HMGB1) on DRibbles was crucial for inducing human B cells activation. Therefore, these findings provide further evidence to promote the clinical application of B-DRibbles vaccines.


Assuntos
Antígenos de Neoplasias/imunologia , Autofagossomos/fisiologia , Linfócitos B/imunologia , Memória Imunológica , Ativação Linfocitária , Linfócitos T/imunologia , Linhagem Celular Tumoral , Proteína HMGB1/fisiologia , Humanos
8.
Int J Radiat Oncol Biol Phys ; 111(2): 491-501, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34044094

RESUMO

PURPOSE: Radiation therapy, which is vital for the treatment of primary liver cancer, comes with unavoidable liver injury, which limits its implementation. N6-methyladenosine (m6A) methylation is involved in many molecular functions. However, its role in radiation-induced liver diseases (RILD) remains unknown. Herein, we investigate the role of m6A methylation in RILD. METHODS AND MATERIALS: Methylated RNA-immunoprecipitation sequencing and RNA transcriptome sequencing were used to reveal the methylation pattern of human hepatic stellate cells (HSCs) exposed to irradiation. C3H/HeN mice and stimulator of interferon genes (STING)-deficient mice underwent x-ray irradiation of 24 Gy in 3 fractions. The m6A methylation of the high-mobility group box 1 (HMGB1) transcript was validated using methylated RNA immunoprecipitation, RNA immunoprecipitation, luciferase assays, and a messenger RNA decay assay. RESULTS: Human hepatic stellate cells showed significant differences in methylation patterns after 8 Gy of x-ray irradiation. Irradiation recruited AlkB homolog 5 (ALKBH5) to demethylate m6A residues in the 3' untranslated region of HMGB1, which resulted in the activation of STING-interferon regulatory factor 3 signaling. Changes in the transcription of the 3' untranslated region of HMGB1 occurred after the knockdown of ALKBH5, which were eliminated after m6A residue mutation. Strikingly, ALKBH5 deficiency or HMGB1 silencing both attenuated type I interferon production and decreased hepatocyte apoptosis. In vivo depletion of ALKBH5 abolished the upregulation of HMGB1-mediated STING signaling and decreased liver inflammation, which was consistent with STING-/- mice treated with irradiation. Notably, YTHDF2 (m6A reader protein) directly bound to HMGB1 m6A-modified sites and promoted its degradation. CONCLUSIONS: ALKBH5-dependent HMGB1 expression mediates STING-interferon regulatory factor 3 innate immune response in RILD.


Assuntos
Homólogo AlkB 5 da RNA Desmetilase/fisiologia , Proteína HMGB1/fisiologia , Células Estreladas do Fígado/efeitos da radiação , Imunidade Inata/efeitos da radiação , Hepatopatias/etiologia , Neoplasias Hepáticas/radioterapia , Proteínas de Membrana/fisiologia , Animais , Proteína HMGB1/genética , Humanos , Interferon Tipo I/biossíntese , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Proteínas de Ligação a RNA/fisiologia , Transdução de Sinais/fisiologia
9.
Int J Mol Sci ; 22(7)2021 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-33807275

RESUMO

High mobility group box 1 (HMGB1) has been demonstrated to promote the migration and invasion of non-small cell lung cancer (NSCLC). However, the mechanism of action of HMGB1 in regulating tumor mobility remains unclear. Therefore, we aimed to investigate whether HMGB1 affects mitochondria distribution and regulates dynamin-related protein 1 (DRP1)-mediated lamellipodia/filopodia formation to promote NSCLC migration. The regulation of mitochondrial membrane tension, dynamics, polarization, fission process, and cytoskeletal rearrangements in lung cancer cells by HMGB1 was analyzed using confocal microscopy. The HMGB1-mediated regulation of DRP1 phosphorylation and colocalization was determined using immunostaining and co-immunoprecipitation assays. The tumorigenic potential of HMGB1 was assessed in vivo and further confirmed using NSCLC patient samples. Our results showed that HMGB1 increased the polarity and mobility of cells (mainly by regulating the cytoskeletal system actin and microtubule dynamics and distribution), promoted the formation of lamellipodia/filopodia, and enhanced the expression and phosphorylation of DRP1 in both the nucleus and cytoplasm. In addition, HMGB1 and DRP1 expressions were positively correlated and exhibited poor prognosis and survival in patients with lung cancer. Collectively, HMGB1 plays a key role in the formation of lamellipodia and filopodia by regulating cytoskeleton dynamics and DRP1 expression to promote lung cancer migration.


Assuntos
Dinaminas/metabolismo , Proteína HMGB1/metabolismo , Neoplasias Pulmonares/metabolismo , Células A549 , Animais , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Linhagem Celular Tumoral , Movimento Celular/genética , Dinaminas/fisiologia , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Proteínas HMGB/metabolismo , Proteína HMGB1/fisiologia , Humanos , Neoplasias Pulmonares/genética , Masculino , Camundongos , Camundongos SCID , Microscopia Confocal/métodos , Mitocôndrias/genética , Dinâmica Mitocondrial , Proteínas Mitocondriais/metabolismo , Fosforilação , Pseudópodes/metabolismo
10.
Mediators Inflamm ; 2021: 5597118, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33859538

RESUMO

Obesity is a well-known public health issue around the world. Sepsis is a lethal clinical syndrome that causes multiorgan failure. Obesity may aggravate inflammation in septic patients. Glutamine (GLN) is a nutrient with immune regulatory and anti-inflammatory properties. Since sepsis is a common contributing factor for acute kidney injury (AKI), this study investigated the effects of GLN administration on sepsis-induced inflammation and AKI in obese mice. A high-fat diet which consists of 60% of calories from fat was provided for 10 weeks to induce obesity in the mice. Then, the obese mice were subdivided into sepsis with saline (SS) or GLN (SG) groups. Cecal ligation and puncture (CLP) was performed to produce sepsis. The SS group was intravenously injected with saline while the SG group was administered GLN one or two doses after CLP. Obese mice with sepsis were sacrificed at 12, 24, or 48 h post-CLP. Results revealed that sepsis resulted in upregulated high-mobility group box protein-1 pathway-associated gene expression in obese mice. Also, expressions of macrophage/neutrophil infiltration markers and inflammatory cytokines in kidneys were elevated. Obese mice treated with GLN after sepsis reversed the depletion of plasma GLN, reduced production of lipid peroxides, and downregulated macrophage/neutrophil infiltration and the inflammatory-associated pathway whereas tight junction gene expression increased in the kidneys. These findings suggest that intravenously administered GLN to obese mice after sepsis alleviated inflammation and attenuated AKI. This model may have clinical application to obese patients with a risk for infection in abdominal surgery.


Assuntos
Injúria Renal Aguda/tratamento farmacológico , Glutamina/uso terapêutico , Inflamação/tratamento farmacológico , Obesidade/complicações , Sepse/complicações , Injúria Renal Aguda/metabolismo , Aminoácidos/sangue , Animais , Dieta Hiperlipídica , Proteína HMGB1/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos
11.
J Mol Med (Berl) ; 99(3): 403-414, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33409553

RESUMO

Acute myeloid leukemia (AML) is an aggressive hematologic malignancy with poor prognosis and overall survival. Clinical investigations show that chronic stress is commonly present in the course of AML and associated with adverse outcome. However, the underlying molecular mechanisms are elusive. In the present study, a chronic restraint stress mouse model was established to evaluate the effect of stress on AML. We found that mice under chronic stress exhibited significantly increased liver and spleen infiltration of leukemic cells and poorer overall survival. This was accompanied by elevated cellular NLR family pyrin domain containing 3 (NLRP3) and interleukin-1ß (IL-1ß) in the liver or bone marrow, and secreted IL-1ß in the plasma, indicating the activation of inflammasomes under chronic restraint stress. High mobility group box 1 (HMGB1) expression was markedly increased in newly diagnosed AML patients, but reduced in complete remission AML patients. The expression level of HMGB1 was positively correlated with NLRP3 mRNA in AML patients. Knockdown of HMGB1 significantly decreased NLRP3 and IL-1ß expression in AML cell lines, and secreted IL-1ß in supernatant of AML cell culture, while HMGB1 stimulation caused contrary effects. These results implied that HMGB1 could be involved in the regulation of inflammasome activation in AML development. Mice model showed that chronic restraint stress-facilitated proliferation and infiltration of AML cells were largely abrogated by knocking down HMGB1. Knockdown of HMGB1 also ameliorated overall survival and remarkably neutralized NLRP3 and IL-1ß expression under chronic restraint stress. These findings provide evidences that chronic stress promotes AML progression via HMGB1/NLRP3/IL-1ß dependent mechanism, suggesting that HMGB1 is a potential therapeutic target for AML. KEY MESSAGES: • Chronic restraint stress promoted acute myeloid leukemia (AML) progression and mediated NLRP3 inflammasome activation in xenograft mice. • HMGB1 mediated NLRP3 inflammasome activation in AML cells. • Knockdown of HMGB1 inhibited AML progression under chronic stress in vivo.


Assuntos
Regulação Leucêmica da Expressão Gênica , Proteína HMGB1/fisiologia , Interleucina-1beta/fisiologia , Leucemia Mieloide Aguda/fisiopatologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Proteínas de Neoplasias/fisiologia , Transdução de Sinais/fisiologia , Animais , Medula Óssea/metabolismo , Linhagem Celular Tumoral , Doença Crônica , Progressão da Doença , Feminino , Técnicas de Silenciamento de Genes , Proteína HMGB1/antagonistas & inibidores , Proteína HMGB1/biossíntese , Proteína HMGB1/genética , Xenoenxertos , Humanos , Inflamassomos/metabolismo , Inflamação , Interleucina-1beta/biossíntese , Interleucina-1beta/genética , Leucemia Mieloide Aguda/metabolismo , Fígado/metabolismo , Fígado/patologia , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR/biossíntese , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Interferência de RNA , Indução de Remissão , Restrição Física , Baço/metabolismo , Baço/patologia , Estresse Fisiológico , Receptor 4 Toll-Like/fisiologia
12.
Int J Mol Sci ; 21(21)2020 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-33158052

RESUMO

The cross-talk between apoptosis and autophagy influences anticancer drug sensitivity and cellular death in various cancer cell lines. However, the fundamental mechanisms behind this phenomenon are still unidentified. We demonstrated anti-cancerous role of cisplatin (CP) and morin hydrate (Mh) as an individual and/or in combination (CP-Mh) in hepatoma cells and tumor model. Exposure of CP resulted in the production of intracellular reactive oxygen species (ROS)-mediated cellular vacuolization, expansion of mitochondria membrane and activation of endoplasmic reticulum (ER)-stress. Consequently, Cyt c translocation led to the increase of Bax/Bcl-2 ratio, which simultaneously triggered caspase-mediated cellular apoptosis. In addition, CP-induced PARP-1 activation led to ADP-ribosylation of HMGB1, which consequently developed autophagy as evident by the LC3I/II ratio. Chemically-induced inhibition of autophagy marked by increased cell death signified a protective role of autophagy against CP treatment. CP-Mh abrogates the PARP-1 expression and significantly reduced HMGB1-cytoplasmic translocation with subsequent inhibition of the HMGB1-Beclin1 complex formation. In the absence of PARP-1, a reduced HMGB1 mediated autophagy was observed followed by induced caspase-dependent apoptosis. To confirm the role of PARP-1-HMGB1 signaling in autophagy, we used the PARP-1 inhibitor, 4-amino-1,8-naphthalimide (ANI), HMGB1 inhibitor, ethyl pyruvate (EP), autophagy inhibitors, 3-methyl adenine (3-MA) and bafilomycin (baf) and small interfering RNAs (siRNA) to target Atg5 in combination of CP and Mh. Exposure to these inhibitors enhanced the sensitivity of HepG2 cells to CP. Collectively, our findings indicate that CP-Mh in combination served as a prominent regulator of autophagy and significant inducer of apoptosis that maintains a homeostatic balance towards HepG2 cells and the subcutaneous tumor model.


Assuntos
Autofagia/efeitos dos fármacos , Carcinoma Hepatocelular/patologia , Cisplatino/farmacologia , Flavonoides/farmacologia , Neoplasias Hepáticas/patologia , Animais , Autofagia/genética , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Cisplatino/administração & dosagem , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Sinergismo Farmacológico , Quimioterapia Combinada , Flavonoides/administração & dosagem , Proteína HMGB1/fisiologia , Células Hep G2 , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Poli(ADP-Ribose) Polimerase-1/fisiologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Biochem Biophys Res Commun ; 532(4): 640-646, 2020 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-32912629

RESUMO

Endothelial cells injury and pro-inflammation cytokines release are the initial steps of hyperhomocysteinemia (HHcy)-associated vascular inflammation. Pyroptosis is a newly identified pro-inflammation form of programmed cell death, causing cell lysis and IL-1ß release, and characterized by the caspases-induced cleavage of its effector molecule gasdermins (GSDMs). However, the effect of homocysteine (Hcy) on endothelial cells pyroptosis and the underlying mechanisms have not been fully defined. We have previously reported that Hcy induces vascular endothelial inflammation accompanied by the increase of high mobility group box-1 protein (HMGB1) and lysosomal cysteine protease cathepsin V in endothelial cells, and other studies have shown that HMGB1 or cathepsins are involved in activation of NLRP3 inflammasome and caspase-1. Here, we investigated the role of HMGB1 and cathepsin V in the process of Hcy-induced pyroptosis. We observed an increase in plasma IL-1ß levels in HHcy patients and mice models, cathepsin V inhibitor reduced the plasma IL-1ß levels and cleavage of GSDMD full-length into GSDMD N-terminal in the thoracic aorta of hyperhomocysteinemia mice. Using cultured HUVECs, we observed that Hcy promoted GSDMD N-terminal expression, silencing GSDMD or HMGB1 rescued Hcy-induced pyroptosis. HMGB1 also increased GSDMD N-terminal expression, and silencing cathepsin V reversed HMGB1-induced pyroptosis. HMGB1 could increase lysosome permeability, and silencing cathepsin V attenuated HMGB1-induced activation of caspase-1. In conclusion, this study has delineated a novel mechanism that HMGB1 mediated Hcy-induced endothelial cells pyroptosis partly via cathepsin V-dependent pathway.


Assuntos
Catepsinas/fisiologia , Cisteína Endopeptidases/fisiologia , Endotélio Vascular/citologia , Proteína HMGB1/fisiologia , Homocisteína/fisiologia , Piroptose , Idoso , Animais , Caspase 1/metabolismo , Linhagem Celular , Feminino , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Hiper-Homocisteinemia/sangue , Hiper-Homocisteinemia/metabolismo , Interleucina-1beta/sangue , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Proteínas de Ligação a Fosfato/metabolismo , Artérias Torácicas/metabolismo
14.
Cell Immunol ; 356: 104192, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32853967

RESUMO

BACKGROUND: CD4+CD25+FoxP3+ T helper cells (Tregs), a subgroup of CD4+ T helper cells, are critical effectors that protect against acute lung injury (ALI) by contact-dependent suppression or releasing anti-inflammatory cytokines including interleukin-10 (IL-10), and transforming growth factor (TGF-ß). HMGB1 (High mobility group box 1 protein) was identified as a nuclear non-histone DNA-binding chromosomal protein, which participates in the regulation of lung inflammatory response and pathological processes in ALI. Previous studies have suggested that Tregs overexpresses the HMGB1-recognizing receptor. However, the interaction of HMGB1 with Tregs in ALI is still unclear. OBJECTIVE: To investigate whether HMGB1 aggravates ALI by suppressing immunosuppressive function of Tregs. METHODS: Anti-HMGB1 antibody and recombinant mouse HMGB1 (rHMGB1) were administered in lipopolysaccharide (LPS)-induced ALI mice and polarized LPS-primed Tregs in vitro. The Tregs pre-stimulated with or without rHMGB1 were adoptively transferred to ALI mice and depleted by Diphtheria toxin (DT). For coculture experiment, isolated Tregs were first pre-stimulated with or without rHMGB1 or anti-HMGB1 antibody, then they were cocultured with bone marrow-derived macrophages (BMMs) under LPS stimulation. RESULTS: Tregs protected against acute lung pathological injury. HMGB1 modulated the suppressive function of Tregs as follows: reduction in the number of the cells and the activity of Tregs, the secretion of anti-inflammatory cytokines (IL-10, TGF-ß) from Tregs, the production of IL-2 from CD4+ T cells and CD11c+ DCs, and the M2 polarization of macrophages, as well as inducing proinflammatory response of macrophages. CONCLUSIONS: HMGB1 could aggravate LPS induced-ALI through suppressing the activity and function of Tregs.


Assuntos
Lesão Pulmonar Aguda/imunologia , Proteína HMGB1/imunologia , Linfócitos T Reguladores/imunologia , Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar Aguda/patologia , Animais , Citocinas/metabolismo , Modelos Animais de Doenças , Proteína HMGB1/metabolismo , Proteína HMGB1/fisiologia , Interleucina-10/imunologia , Lipopolissacarídeos/farmacologia , Pulmão/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T Reguladores/metabolismo , Fator de Crescimento Transformador beta/metabolismo
15.
J Hematol Oncol ; 13(1): 116, 2020 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-32831115

RESUMO

High mobility group box 1 (HMGB1) is a non-histone chromatin-associated protein widely distributed in eukaryotic cells and is involved in DNA damage repair and genomic stability maintenance. In response to stimulus like bacteria or chemoradiotherapy, HMGB1 can translocate to extracellular context as a danger alarmin, activate the immune response, and participate in the regulation of inflammation and cancer progression.


Assuntos
Proteína HMGB1/fisiologia , Inflamação/metabolismo , Proteínas de Neoplasias/fisiologia , Neoplasias/metabolismo , Apoptose/fisiologia , Autofagia/fisiologia , Carcinogênese , Núcleo Celular/metabolismo , Sobrevivência Celular/fisiologia , Quimiorradioterapia , Citocinas/metabolismo , Citoplasma/metabolismo , Reparo do DNA , Humanos , Imunidade/fisiologia , Imunoterapia , Neoplasias/terapia
16.
J Hematol Oncol ; 13(1): 91, 2020 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-32660524

RESUMO

High mobility group box 1 (HMGB1) is a nonhistone chromatin-associated protein that has been widely reported to play a pivotal role in the pathogenesis of hematopoietic malignancies. As a representative damage-associated molecular pattern (DAMP), HMGB1 normally exists inside cells but can be secreted into the extracellular environment through passive or active release. Extracellular HMGB1 binds with several different receptors and interactors to mediate the proliferation, differentiation, mobilization, and senescence of hematopoietic stem cells (HSCs). HMGB1 is also involved in the formation of the inflammatory bone marrow (BM) microenvironment by activating proinflammatory signaling pathways. Moreover, HMGB1-dependent autophagy induces chemotherapy resistance in leukemia and multiple myeloma. In this review, we systematically summarize the emerging roles of HMGB1 in carcinogenesis, progression, prognosis, and potential clinical applications in different hematopoietic malignancies. In summary, targeting the regulation of HMGB1 activity in HSCs and the BM microenvironment is highly beneficial in the diagnosis and treatment of various hematopoietic malignancies.


Assuntos
Proteína HMGB1/fisiologia , Neoplasias Hematológicas/metabolismo , Proteínas de Neoplasias/fisiologia , Antineoplásicos/uso terapêutico , Autofagia , Biomarcadores Tumorais , Medula Óssea/patologia , Progressão da Doença , Resistencia a Medicamentos Antineoplásicos , Líquido Extracelular/metabolismo , Proteína HMGB1/química , Proteína HMGB1/imunologia , Neoplasias Hematológicas/diagnóstico , Neoplasias Hematológicas/terapia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/patologia , Humanos , Inflamação , Leucemia/diagnóstico , Leucemia/metabolismo , Leucemia/terapia , Linfoma/diagnóstico , Linfoma/metabolismo , Linfoma/terapia , Mieloma Múltiplo/diagnóstico , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/terapia , Síndromes Mielodisplásicas/diagnóstico , Síndromes Mielodisplásicas/metabolismo , Síndromes Mielodisplásicas/terapia , Proteínas de Neoplasias/química , Proteínas de Neoplasias/imunologia , Células-Tronco Neoplásicas/patologia , Oxirredução , Processamento de Proteína Pós-Traducional , Transdução de Sinais/fisiologia , Microambiente Tumoral
17.
Rhinology ; 58(5): 495-505, 2020 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-32478338

RESUMO

BACKGROUND: In the treatment of rhinosinusitis, nasal polyps are a major problem, and the epithelial-to-mesenchymal transition (EMT) process is considered pivotal in their development. Although various studies have addressed the role of high mobility group box 1 (HMGB1) nuclear protein in this setting, its impact on EMT has yet to be evaluated. Our aim was the pathogenic mechanism of HMGB1 in EMT and EMT-induced upper respiratory nasal polyps. METHODS: We investigated the EMT-related effects of HMGB1 in human nasal epithelial (HNE) cells using western blot analysis, transepithelial-electrical resistance (TEER) testing, wound healing assay, and immunofluorescence. HNE cells were incubated in a low-oxygen environment to evaluate the role of HMGB1 in hypoxia-induced EMT. Further support for our in vitro findings was obtained through murine models. Human nasal polyps and nasal lavage fluid samples were collected for western blotting, immunohistochemistry, and enzyme-linked immunosorbent assay (ELISA). RESULTS: HMGB1 increased mesenchymal markers and decreased epithelial markers in HNE cells. Hypoxia-induced HMGB1 in turn induced EMT, apparently through RAGE signaling. We verified HMGB1-induced EMT in the upper respiratory epithelium of mice by instilling intranasal HMGB1. In testing of human nasal polyps, HMGB1 and mesenchymal markers were heightened, whereas epithelial markers were reduced, compared with tissue controls. CONCLUSION: HMGB1 secretion in nasal epithelium may be a major pathogenic factor in upper respiratory EMT, contributing to nasal polyps.


Assuntos
Proteína HMGB1 , Pólipos Nasais , Sinusite , Animais , Células Epiteliais , Transição Epitelial-Mesenquimal , Proteína HMGB1/metabolismo , Proteína HMGB1/fisiologia , Humanos , Camundongos , Pólipos Nasais/metabolismo , Sinusite/metabolismo
18.
Cell Prolif ; 53(6): e12829, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32419317

RESUMO

OBJECTIVE: Acute kidney injury (AKI) is a common complication of acute liver failure (ALF). Pyroptosis is a necrosis type related to inflammation. This study aimed to investigate the role of TNF-α/HMGB1 pathway in pyroptosis during ALF and AKI. METHODS: An ALF and AKI mouse model was generated using LPS/D-Gal, and a TNF-α inhibitor, CC-5013, was used to treat the mice. THP-1 cells were induced to differentiate into M1 macrophages, then challenged with either CC-5013 or an HMGB1 inhibitor, glycyrrhizin. pLVX-mCMVZsGreen-PGK-Puros plasmids containing TNF-α wild-type (WT), mutation A94T of TNF-α and mutation P84L of TNF-α were transfected into M1 macrophages. RESULTS: Treatment with CC-5013 decreased the activation of TNF-α/HMGB1 pathway and pyroptosis in the treated mice and cells compared with the control mice and cells. CC-5013 also ameliorated liver and kidney pathological changes and improved liver and renal functions in treated mice, and the number of M1 macrophages in the liver and kidney tissues also decreased. The activation of TNF-α/HMGB1 pathway and pyroptosis increased in the M1 macrophage group compared with the normal group. Similarly, the activation of TNF-α/HMGB1 pathway and pyroptosis in the LPS + WT group also increased. By contrast, the activation of the TNF-α/HMGB1 pathway and pyroptosis decreased in the LPS + A94T and LPS + P84L groups. Moreover, glycyrrhizin inhibited pyroptosis. CONCLUSION: The TNF-α/HMGB1 inflammation signalling pathway plays an important role in pyroptosis during ALF and AKI.


Assuntos
Injúria Renal Aguda/metabolismo , Proteína HMGB1/fisiologia , Falência Hepática Aguda/metabolismo , Piroptose , Fator de Necrose Tumoral alfa/fisiologia , Injúria Renal Aguda/sangue , Injúria Renal Aguda/imunologia , Injúria Renal Aguda/patologia , Animais , Diferenciação Celular/efeitos dos fármacos , Ácido Glicirrízico/farmacologia , Proteína HMGB1/antagonistas & inibidores , Proteína HMGB1/sangue , Proteína HMGB1/metabolismo , Humanos , Inflamação/metabolismo , Lenalidomida/farmacologia , Falência Hepática Aguda/sangue , Falência Hepática Aguda/imunologia , Falência Hepática Aguda/patologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Polimorfismo de Nucleotídeo Único , Piroptose/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Células THP-1 , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/genética
19.
J Pharmacol Sci ; 143(2): 112-116, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32222337

RESUMO

HMGB1, a nuclear protein, once released to the extracellular space, promotes somatic and visceral pain signals. We thus analyzed the role of HMGB1 in an intravesical substance P-induced bladder pain syndrome (BPS) mouse model. Intravesical administration of substance P caused referred hyperalgesia/allodynia in the lower abdomen and hindpaw without producing severe urothelial damage, which was prevented by an anti-HMGB1-neutralizing antibody, thrombomodulin α capable of inactivating HMGB1 and antagonists of RAGE or CXCR4. The HMGB1 inactivation or RAGE blockade also reversed the established bladder pain symptoms. HMGB1 and RAGE are thus considered to serve as therapeutic targets for BPS.


Assuntos
Anticorpos Neutralizantes/uso terapêutico , Cistite Intersticial/etiologia , Cistite Intersticial/genética , Proteína HMGB1/fisiologia , Receptores Citoplasmáticos e Nucleares , Substância P/efeitos adversos , Trombomodulina/uso terapêutico , Animais , Cistite Intersticial/tratamento farmacológico , Modelos Animais de Doenças , Feminino , Proteína HMGB1/imunologia , Humanos , Masculino , Camundongos Endogâmicos , Terapia de Alvo Molecular , Receptor para Produtos Finais de Glicação Avançada/antagonistas & inibidores , Receptores CXCR4/antagonistas & inibidores , Substância P/administração & dosagem
20.
Life Sci ; 246: 117415, 2020 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-32035932

RESUMO

High-mobility group box 1 protein (HMGB1) is a crucial proinflammatory cytokine that contributes to acute lung injury (ALI). Macrophages are known to express the primary receptors (Toll-like receptor [TLR] 2, and TLR4) of HMGB1 for transmitting intracellular signals. Studies have revealed that double-stranded RNA activated protein kinase R (PKR), which is expressed in macrophages, participates in ALI by regulating macrophage polarization and proinflammatory cytokine release, and that PKR is normally activated by a subset of TLRs. The present study investigated whether HMGB1 engages in ALI by activating PKR in macrophages and inducing classically activated macrophage (M1) polarization via TLR2- and TLR4-mediated nuclear factor (NF)-κB signaling pathways. In an vivo mouse model of lipopolysaccharide (LPS)-induced ALI, anti-HMGB1, rHMGB1, LPS-RS (TLR2 and TLR4 antagonist), or C16 (PKR inhibitor) was administered to mice 2 h after LPS challenge or 1 h before LPS challenge. In vitro, bone marrow-derived macrophages from mice primed with LPS were stimulated with or without anti-HMGB1, rHMGB1, LPS-RS, or C16. Our studies revealed that rHMGB1 stimulation induced M1 polarization in ALI, and that anti-HMGB1 and C16 treatments had the opposite effect. Anti-HMGB1 and LPS-RS significantly inhibited LPS-induced PKR expression in macrophages; however, rHMGB1 administration increased PKR expression. These results indicate that HMGB1 participates in the pathogenesis of ALI by activating PKR in macrophages and inducing M1 polarization through TLR2- and TLR4-mediated NF-κB signaling pathways.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Proteína HMGB1/fisiologia , Ativação de Macrófagos , eIF-2 Quinase/metabolismo , Lesão Pulmonar Aguda/etiologia , Animais , Western Blotting , Modelos Animais de Doenças , Ativação Enzimática , Ensaio de Imunoadsorção Enzimática , Proteína HMGB1/metabolismo , Interleucinas/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase em Tempo Real , Proteínas da Matriz Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA