Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cells ; 10(8)2021 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-34440779

RESUMEN

After retinal detachment (RD), the induction of autophagy protects photoreceptors (PR) from apoptotic cell death. The cytoplasmic high-mobility group box 1 (HMGB1) promotes autophagy. We previously demonstrated that the deletion of HMGB1 from rod PRs results in a more rapid death of these cells after RD. In this work, we tested the hypothesis that the lack of HMGB1 accelerates PR death after RD due to the reduced activation of protective autophagy in the retina after RD. The injection of 1% hyaluronic acid into the subretinal space was used to create acute RD in mice with a rhodopsin-Cre-mediated conditional knockout (cKO) of HMGB1 in rods (HMGB1Δrod) and littermate controls. RD sharply increased the number of apoptotic cells in the outer nuclear layer (ONL), and this number was further increased in HMGB1Δrod mouse retinas. The activation of autophagy after RD was reduced in the HMGB1Δrod mouse retinas compared to controls, as evidenced by diminished levels of autophagy regulatory proteins LC3-II, Beclin1, ATG5/12, and phospho-ATG16L1. The cKO of HMGB1 in rods increased the expression of Fas and the Bax/Bcl-2 ratio in detached retinas, promoting apoptotic cell death. In conclusion, endogenous HMGB1 facilitates autophagy activation in PR cells following RD to promote PR cell survival and reduce programmed apoptotic cell death.


Asunto(s)
Apoptosis , Autofagia , Proteína HMGB1/deficiencia , Desprendimiento de Retina/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Relacionadas con la Autofagia/metabolismo , Supervivencia Celular , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Proteína HMGB1/genética , Mediadores de Inflamación/metabolismo , Ratones Noqueados , Desprendimiento de Retina/genética , Desprendimiento de Retina/patología , Células Fotorreceptoras Retinianas Bastones/patología , Transducción de Señal
2.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-33443188

RESUMEN

Dysregulation of inflammatory cytokines in keratinocytes promote the pathogenesis of the skin inflammation, such as allergic contact dermatitis (ACD). High-mobility group box 1 protein (HMGB1) has been implicated in the promotion of skin inflammation upon its extracellular release as a damage-associated molecular pattern molecule. However, whether and how HMGB1 in keratinocytes contributes to ACD and other skin disorders remain elusive. In this study, we generated conditional knockout mice in which the Hmgb1 gene is specifically deleted in keratinocytes, and examined its role in ACD models. Interestingly, the mutant mice showed exacerbated skin inflammation, accompanied by increased ear thickening in 2,4-dinitrofluorobenezene-induced ACDs. The mRNA expression of interleukin-24 (IL-24), a cytokine known to critically contribute to ACD pathogenesis, was elevated in skin lesions of the mutant mice. As with constitutively expressed, IL-4-induced Il24 mRNA, expression was also augmented in the Hmgb1-deficient keratinocytes, which would account for the exacerbation of ACD in the mutant mice. Mechanistically, we observed an increased binding of trimethyl histone H3 (lys4) (H3K4me3), a hallmark of transcriptionally active genes, to the promoter region of the Il24 gene in the hmgb1-deficient cells. Thus, the nuclear HMGB1 is a critical "gate keeper" in that the dermal homeostasis is contingent to its function in chromatin remodeling. Our study revealed a facet of nuclear HMGB1, namely its antiinflammatory function in keratinocytes for the skin homeostasis.


Asunto(s)
Ensamble y Desensamble de Cromatina , Dermatitis Alérgica por Contacto/metabolismo , Proteína HMGB1/metabolismo , Histonas/metabolismo , Interleucinas/metabolismo , Queratinocitos/metabolismo , Animales , Dermatitis Alérgica por Contacto/genética , Dermatitis Alérgica por Contacto/prevención & control , Dinitrofluorobenceno/toxicidad , Modelos Animales de Enfermedad , Oído/patología , Eliminación de Gen , Regulación de la Expresión Génica/genética , Proteína HMGB1/deficiencia , Proteína HMGB1/genética , Inflamación/genética , Inflamación/metabolismo , Interleucina-4/farmacología , Interleucinas/genética , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas , Piel/inmunología , Piel/metabolismo , Piel/patología , Quimera por Trasplante
3.
Arterioscler Thromb Vasc Biol ; 41(1): 200-216, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33054399

RESUMEN

OBJECTIVE: LDL (low-density lipoprotein) transcytosis across the endothelium is performed by the SR-BI (scavenger receptor class B type 1) receptor and contributes to atherosclerosis. HMGB1 (high mobility group box 1) is a structural protein in the nucleus that is released by cells during inflammation; extracellular HMGB1 has been implicated in advanced disease. Whether intracellular HMGB1 regulates LDL transcytosis through its nuclear functions is unknown. Approach and Results: HMGB1 was depleted by siRNA in human coronary artery endothelial cells, and transcytosis of LDL was measured by total internal reflection fluorescence microscopy. Knockdown of HMGB1 attenuated LDL transcytosis without affecting albumin transcytosis. Loss of HMGB1 resulted in reduction in SR-BI levels and depletion of SREBP2 (sterol regulatory element-binding protein 2)-a transcription factor upstream of SR-BI. The effect of HMGB1 depletion on LDL transcytosis required SR-BI and SREBP2. Overexpression of HMGB1 caused an increase in LDL transcytosis that was unaffected by inhibition of extracellular HMGB1 or depletion of RAGE (receptor for advanced glycation endproducts)-a cell surface receptor for HMGB1. The effect of HMGB1 overexpression on LDL transcytosis was prevented by knockdown of SREBP2. Loss of HMGB1 caused a reduction in the half-life of SREBP2; incubation with LDL caused a significant increase in nuclear localization of HMGB1 that was dependent on SR-BI. Animals lacking endothelial HMGB1 exhibited less acute accumulation of LDL in the aorta 30 minutes after injection and when fed a high-fat diet developed fewer fatty streaks and less atherosclerosis. CONCLUSIONS: Endothelial HMGB1 regulates LDL transcytosis by prolonging the half-life of SREBP2, enhancing SR-BI expression. Translocation of HMGB1 to the nucleus in response to LDL requires SR-BI.


Asunto(s)
Aterosclerosis/metabolismo , Células Endoteliales/metabolismo , Proteína HMGB1/metabolismo , Receptores de LDL/metabolismo , Receptores Depuradores de Clase B/metabolismo , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo , Transcitosis , Transporte Activo de Núcleo Celular , Animales , Aterosclerosis/genética , Aterosclerosis/patología , Aterosclerosis/prevención & control , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Proteína HMGB1/deficiencia , Proteína HMGB1/genética , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Estabilidad Proteica , Receptores de LDL/genética , Receptores Depuradores de Clase B/genética , Transducción de Señal , Proteína 2 de Unión a Elementos Reguladores de Esteroles/genética
4.
Front Immunol ; 11: 1122, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32670275

RESUMEN

Acute inflammation is a complex biological response of tissues to harmful stimuli, such as pathogens or cell damage, and is essential for immune defense and proper healing. However, unresolved inflammation can lead to chronic disorders, including cancer and fibrosis. The High Mobility Group Box 1 (HMGB1) protein is a Damage-Associated Molecular Pattern (DAMP) molecule that orchestrates key events in inflammation by switching among mutually exclusive redox states. Fully reduced HMGB1 (frHMGB1) supports immune cell recruitment and tissue regeneration, while the isoform containing a disulphide bond (dsHMGB1) promotes secretion of inflammatory mediators by immune cells. Although it has been suggested that the tissue itself determines the redox state of the extracellular space and of released HMGB1, the dynamics of HMGB1 oxidation in health and disease are unknown. In the present work, we analyzed the expression of HMGB1 redox isoforms in different inflammatory conditions in skeletal muscle, from acute injury to muscle wasting, in tumor microenvironment, in spleen, and in liver after drug intoxication. Our results reveal that the redox modulation of HMGB1 is tissue-specific, with high expression of dsHMGB1 in normal spleen and liver and very low in muscle, where it appears after acute damage. Similarly, dsHMGB1 is highly expressed in the tumor microenvironment while it is absent in cachectic muscles from the same tumor-bearing mice. These findings emphasize the accurate and dynamic regulation of HMGB1 redox state, with the presence of dsHMGB1 tightly associated with leukocyte infiltration. Accordingly, we identified circulating, infiltrating, and resident leukocytes as reservoirs and transporters of dsHMGB1 in tissue and tumor microenvironment, demonstrating that the redox state of HMGB1 is controlled at both tissue and cell levels. Overall, our data point out that HMGB1 oxidation is a timely and spatially regulated process in physiological and pathological conditions. This precise modulation might play key roles to finetune inflammatory and regenerative processes.


Asunto(s)
Proteína HMGB1/metabolismo , Animales , Caquexia/inmunología , Caquexia/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/inmunología , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Modelos Animales de Enfermedad , Proteína HMGB1/deficiencia , Proteína HMGB1/inmunología , Inflamación/inmunología , Inflamación/metabolismo , Leucocitos/inmunología , Leucocitos/metabolismo , Hígado/inmunología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/inmunología , Músculo Esquelético/lesiones , Músculo Esquelético/metabolismo , Oxidación-Reducción , Bazo/inmunología , Bazo/metabolismo , Microambiente Tumoral/inmunología , Microambiente Tumoral/fisiología
5.
Cell Death Differ ; 27(5): 1489-1504, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31595043

RESUMEN

A reciprocal communication between the implantation-competent blastocyst and the receptive uterus is essential to successful implantation and pregnancy success. Progesterone (P4) signaling via nuclear progesterone receptor (PR) is absolutely critical for pregnancy initiation and its success in most eutherian mammals. Here we show that a nuclear protein high-mobility group box-1 (HMGB1) plays a critical role in implantation in mice by preserving P4-PR signaling. Conditional deletion of uterine Hmgb1 by a Pgr-Cre driver shows implantation defects accompanied by decreased stromal cell Hoxa10 expression and cell proliferation, two known signatures of inefficient responsiveness of stromal cells to PR signaling in implantation. These mice evoke inflammatory conditions with sustained macrophage accumulation in the stromal compartment on day 4 of pregnancy with elevated levels of macrophage attractants Csf1 and Ccl2. The results are consistent with the failure of exogenous P4 administration to rescue implantation deficiency in the mutant females. These early defects are propagated throughout the course of pregnancy and ultimately result in substantial subfertility. Collectively, the present study provides evidence that nuclear HMGB1 contributes to successful blastocyst implantation by sustaining P4-PR signaling and restricting macrophage accumulation to attenuate harmful inflammatory responses.


Asunto(s)
Proteína HMGB1/deficiencia , Útero/metabolismo , Útero/patología , Animales , Citocinas/metabolismo , Decidua/patología , Implantación del Embrión , Femenino , Eliminación de Gen , Proteína HMGB1/metabolismo , Infertilidad Femenina/patología , Macrófagos/metabolismo , Masculino , Ratones , Embarazo , Resultado del Embarazo , Receptores de Progesterona/metabolismo , Células del Estroma/metabolismo
6.
Artif Cells Nanomed Biotechnol ; 47(1): 2967-2971, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31313594

RESUMEN

To study the effects of ginsenoside Rb1 and the molecular mechanisms on proliferation and apoptosis of uterine fibroid cells, Rb1 + pc DNA3.1, Rb1 + pc DNA3.1-HMGB1, si-NC or si-HMGB1 was transfected into uterine fibroid cells by liposome method; the inhibitory rate and proliferation of human uterine fibroid cells were detected by MTT assay; apoptosis of uterine fibroid cells was detected by flow cytometry assay; HMGB1 protein expression in uterine fibroid cells was detected by Western blot assay. Compared with untreated uterine fibroid cells, the inhibitory and apoptosis rate of uterine fibroid cells treated with Rb1 were significantly up-regulated, while the expression level of HMGB1 was significantly down-regulated (p < .05). HMGB1 knockdown inhibited proliferation and promoted apoptosis of uterine fibroid cells. HMGB1 overexpression reversed the inhibitory effect on proliferation and the promotion effect on apoptosis of Rb1 in uterine fibroid cells. Ginsenoside Rb1 could inhibit uterine fibroid cells proliferation and promote apoptosis. This mechanism might be directly related to the downregulation of HMGB1, providing a basis for the treatment of uterine fibroids with ginsenoside Rb1.


Asunto(s)
Apoptosis/efectos de los fármacos , Ginsenósidos/farmacología , Proteína HMGB1/metabolismo , Leiomioma/metabolismo , Leiomioma/patología , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Femenino , Técnicas de Silenciamiento del Gen , Proteína HMGB1/deficiencia , Proteína HMGB1/genética , Humanos , Leiomioma/genética
7.
J Immunol Methods ; 456: 72-76, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29453955

RESUMEN

HMGB1 and HMGB2 are DNA-interacting proteins but can also have extracellular actions during inflammation. Despite their relatively high homology, they may have distinct roles, making it essential to be able to differentiate between the two. Here we examine the specificity of five commercially-available anti-HMGB1 antibodies. By Western blotting of recombinant proteins and HMGB1-/- mouse embryonic fibroblasts, we identified only one HMGB1 antibody that, under our experimental conditions, did not also detect HMGB2. Selecting specific antibodies for HMGB1 and HMGB2 allowed identification of distinct HMGB1 and HMGB2 subcellular pools in primary neutrophils.


Asunto(s)
Anticuerpos/inmunología , Reacciones Cruzadas , Proteína HMGB1/inmunología , Proteína HMGB2/inmunología , Células Endoteliales de la Vena Umbilical Humana/inmunología , Reacciones Antígeno-Anticuerpo , Células Cultivadas , Proteína HMGB1/deficiencia , Proteína HMGB2/deficiencia , Voluntarios Sanos , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos
8.
J Vasc Surg ; 67(3): 910-921, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28259568

RESUMEN

OBJECTIVE: We have previously shown that exogenous administration of the nuclear protein high mobility group box 1 (HMGB1) improves angiogenesis after tissue ischemia. Antagonizing HMGB1 prolongs muscle necrosis and deters regeneration. In this study, we evaluated HMGB1 expression in peripheral arterial disease (PAD) and the mechanisms that promote its release in a murine model of hindlimb ischemia. Specifically, we investigated how chloroquine (CQ), a commonly employed disease-modifying antirheumatic drug, promotes HMGB1 release from muscle. We hypothesized that CQ could increase HMGB1 locally and systemically, allowing it to mediate recovery from ischemic injury. METHODS: Muscle biopsies were performed on patients undergoing lower extremity surgery for non-PAD-related disease as well as for claudication and critical limb ischemia. Clinical symptoms and ankle-brachial indices were recorded for each patient. HMGB1 was detected in muscle sections using immunohistochemical staining. Unilateral femoral artery ligation was performed on both wild-type and inducible HMGB1 knockout mice. Wild-type mice were administered intraperitoneal CQ 2 weeks before and after femoral artery ligation. Laser Doppler perfusion imaging was used to determine perfusion recovery. Serum and tissue levels of HMGB1 were measured at designated time points. In vitro, cultured C2C12 myoblasts were treated with increasing doses of CQ. HMGB1, autophagosome formation, p62/SQSTM1 accumulation, caspase-1 expression and activity, and lactate dehydrogenase levels were measured in supernatants and cell lysates. RESULTS: Nuclear expression of HMGB1 was prominent in patients with claudication and critical limb ischemia (P < .05) compared with controls. CQ-treated mice had elevated serum HMGB1 and diffuse HMGB1 staining in muscle (P < .01). In wild-type mice, CQ treatment resulted in higher laser Doppler perfusion imaging ratios in the ischemic limb at 7 days (P < .03) and less fat replacement after 2 weeks (P < .03). In cultured myoblasts, CQ induced autophagosome accumulation, inhibited p62/SQSTM-1 degradation, and activated caspase-1. CONCLUSIONS: HMGB1 is prominently expressed in PAD muscle but mostly confined to the nucleus. Our in vivo data suggest that HMGB1 mobilization into the sarcoplasm and serum can be increased with CQ, possibly through caspase-1-mediated pathways. Whereas HMGB1 can be released by many cell types, these studies suggest that the muscle may be an important additional source that is relevant in PAD.


Asunto(s)
Cloroquina/farmacología , Arteria Femoral/cirugía , Proteína HMGB1/metabolismo , Claudicación Intermitente/tratamiento farmacológico , Isquemia/tratamiento farmacológico , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/efectos de los fármacos , Enfermedad Arterial Periférica/tratamiento farmacológico , Anciano , Animales , Autofagia/efectos de los fármacos , Velocidad del Flujo Sanguíneo , Estudios de Casos y Controles , Caspasa 1/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Femenino , Proteína HMGB1/deficiencia , Proteína HMGB1/genética , Humanos , Claudicación Intermitente/metabolismo , Claudicación Intermitente/patología , Isquemia/metabolismo , Isquemia/patología , L-Lactato Deshidrogenasa/metabolismo , Ligadura , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Enfermedad Arterial Periférica/metabolismo , Enfermedad Arterial Periférica/patología , Recuperación de la Función , Flujo Sanguíneo Regional , Proteína Sequestosoma-1/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba
9.
Arterioscler Thromb Vasc Biol ; 35(12): 2579-93, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26515416

RESUMEN

OBJECTIVE: Endoluminal vascular interventions such as angioplasty initiate a sterile inflammatory response resulting from local tissue damage. This response drives the development of intimal hyperplasia (IH) that, in turn, can lead to arterial occlusion. We hypothesized that the ubiquitous nuclear protein and damage-associated molecular pattern molecule, high-mobility group box 1 (HMGB1), is one of the endogenous mediators that activates processes leading to IH after endoluminal injury to the arterial wall. The aim of this study is to investigate whether approaches that reduce the levels of HMGB1 or inhibit its activity suppresses IH after arterial injury. APPROACH AND RESULTS: Here, we show that HMGB1 regulates IH in a mouse carotid wire injury model. Induced genetic deletion or neutralization of HMGB1 prevents IH, monocyte recruitment, and smooth muscle cell growth factor production after endoluminal carotid artery injury. A specific inhibitor of HMGB1 myeloid differentiation factor 2-toll-like receptor 4 (TLR4) interaction, P5779, also significantly inhibits IH. HMGB1 deletion is mimicked in this model by global deletion of TLR4 and partially replicated by myeloid-specific deletion of TLR4 but not TLR2 or receptor for advanced glycation endproducts deletion. The specific HMGB1 isoform known to activate TLR4 signaling (disulfide HMGB1) stimulates smooth muscle cell to migrate and produce monocyte chemotactic protein 1/CCL2) via TLR4. Macrophages produce smooth muscle cell mitogens in response to disulfide HMGB1 also in a TLR4/myeloid differentiation primary response gene (88)/Trif-dependent manner. CONCLUSIONS: These findings place HMGB1 and its receptor, TLR4 as critical regulators of the events that drive the inflammation leading to IH after endoluminal arterial injury and identify this pathway as a possible therapeutic target to limit IH to attenuate damage-associated molecular pattern molecule-mediated vascular inflammatory responses.


Asunto(s)
Arterias Carótidas/metabolismo , Traumatismos de las Arterias Carótidas/metabolismo , Proteína HMGB1/metabolismo , Neointima , Receptor Toll-Like 4/metabolismo , Lesiones del Sistema Vascular/metabolismo , Vasculitis/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Arterias Carótidas/patología , Traumatismos de las Arterias Carótidas/genética , Traumatismos de las Arterias Carótidas/patología , Movimiento Celular , Proliferación Celular , Células Cultivadas , Quimiotaxis de Leucocito , Citocinas/metabolismo , Modelos Animales de Enfermedad , Proteína HMGB1/deficiencia , Proteína HMGB1/genética , Humanos , Hiperplasia , Mediadores de Inflamación/metabolismo , Macrófagos Peritoneales/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Transducción de Señal , Factores de Tiempo , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/genética , Lesiones del Sistema Vascular/genética , Lesiones del Sistema Vascular/patología , Vasculitis/genética , Vasculitis/patología
11.
Oncol Res ; 22(5-6): 235-45, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-26629935

RESUMEN

HMGB1, which acts as a DNA chaperone to help maintain nuclear homeostasis, was reported to play a prominent role in cancer progression, angiogenesis, invasion, and metastasis development. Increased expression of HMGB1 has been observed in several tumor entities. However, the molecular mechanisms of HMGB1 in tumorigenesis of bladder cancer have rarely been reported. In the present study, real-time quantitative RT-PCR analysis revealed that the expression of HMGB1 in human bladder urothelial carcinoma (BUC) cells was much higher than that in human normal urethra epithelial cells. In order to investigate the role of HMGB1 in BUC cells, RNA interference and Talen-mediated gene knockout (KO) were used to knockdown and knockout HMGB1, respectively, in BUC cell lines BIU-87 and T24. HMGB1 knockdown/out greatly inhibited proliferation, invasion, and cell cycle G1/S transition of BUC cells. The decrease in cell viability caused by HMGB1 knockdown/out was due to an increase in apoptosis via Bax/Bcl-2, both of which were important molecules involved in the apoptotic pathway. We then investigated the effect of HMGB1 knockdown/out on the sensitivity of BUC cells treated with the anticancer drug cisplatin. Knockdown or knockout of HMGB1 rendered BUC cells more sensitive to cisplatin. The decreased expression of LC3-II and Beclin 1, which resulted in decreased levels of autophagy, could probably explain this phenomenon. Thus, HMGB1 may become a novel promising candidate for the prognosis and therapy for bladder cancer.


Asunto(s)
Proliferación Celular/fisiología , Técnicas de Silenciamiento del Gen , Proteína HMGB1/deficiencia , Neoplasias de la Vejiga Urinaria/metabolismo , Urotelio/metabolismo , Línea Celular Tumoral , Proteína HMGB1/genética , Humanos , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/patología , Urotelio/patología
12.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 29(11): 1159-62, 2013 Nov.
Artículo en Chino | MEDLINE | ID: mdl-24200063

RESUMEN

OBJECTIVE: To investigate the effect of high mobility group-box protein 1 (HMGB1)-siRNA on invasion and migration of human hepatoma cell line HepG2, and further to explore its mechanism. METHODS: HMGB1-siRNA was synthesized with RNA interference and transferred into HepG2 cells to down-regulate the expression of HMGB1. The invasion and migration activities were assayed by Transwell™ assay and monolayer wounding healing assay. The levels of matrix metalloproteinase 2 (MMP-2), MMP-9, intercellular adhesion molecule 1 (ICAM-1) and tissue inhibitor of MMP-2 (TIMP-2) were detected by RT-PCR and Western blotting in HepG2 cells after treatment with 40 nmol/L HMGB1-siRNA for 24 h. RESULTS: The migration and invasion abilities of HepG2 cells were inhibited by 40 nmol/L HMGB1-siRNA markedly. Compared with the control group, MMP-2, MMP-9, ICAM-1 were down-regulated, TIMP-2 was up-regulated significantly by HMGB1-siRNA (P<0.05). CONCLUSION: HMGB1-siRNA can inhibit the invasion and migration abilities of human hepatoma cells by down-regulating the expressions of MMP-2, MMP-9, ICAM-1 and up-regulating the expression of TIMP-2.


Asunto(s)
Carcinoma Hepatocelular/patología , Movimiento Celular , Proteína HMGB1/metabolismo , Neoplasias Hepáticas/patología , Regulación Neoplásica de la Expresión Génica , Proteína HMGB1/deficiencia , Proteína HMGB1/genética , Células Hep G2 , Humanos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Invasividad Neoplásica , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Inhibidor Tisular de Metaloproteinasa-2/genética , Inhibidor Tisular de Metaloproteinasa-2/metabolismo
13.
J Immunol ; 190(4): 1797-806, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23303669

RESUMEN

High-mobility group box 1 protein (HMGB1), which mainly exists in the nucleus, has recently been shown to function as a sentinel molecule for viral nucleic acid sensing and an autophagy regulator in the cytoplasm. In this study, we studied the chaperone-like activity of HMGB1 and found that HMGB1 inhibited the chemically induced aggregation of insulin and lysozyme, as well as the heat-induced aggregation of citrate synthase. HMGB1 also restored the heat-induced suppression of cytoplasmic luciferase activity as a reporter protein in hamster lung fibroblast O23 cells with expression of HMGB1. Next, we demonstrated that HMGB1 inhibited the formation of aggregates and toxicity caused by expanded polyglutamine (polyQ), one of the main causes of Huntington disease. HMGB1 directly interacted with polyQ on immunofluorescence and coimmunoprecipitation assay, whereas the overexpression of HMGB1 or exogenous administration of recombinant HMGB1 protein remarkably reduced polyQ aggregates in SHSY5Y cells and hmgb1(-/-) mouse embryonic fibroblasts upon filter trap and immunofluorescence assay. Finally, overexpressed HMGB1 proteins in mouse embryonic primary striatal neurons also bound to polyQ and decreased the formation of polyQ aggregates. To this end, we have demonstrated that HMGB1 exhibits chaperone-like activity and a possible therapeutic candidate in polyQ disease.


Asunto(s)
Proteína HMGB1/fisiología , Chaperonas Moleculares/metabolismo , Péptidos/antagonistas & inhibidores , Péptidos/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Cricetinae , Cricetulus , Células HEK293 , Proteína HMGB1/deficiencia , Proteína HMGB1/metabolismo , Humanos , Ratones , Ratones Noqueados , Chaperonas Moleculares/química , Chaperonas Moleculares/fisiología , Células 3T3 NIH , Neuroblastoma/metabolismo , Neuroblastoma/terapia
14.
J Immunol ; 187(9): 4686-94, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21957148

RESUMEN

High mobility group box 1 (HMGB1) is a highly conserved protein with multiple intracellular and extracellular functions, including transcriptional regulation, as well as modulation of inflammation, cell migration, and ingestion of apoptotic cells. In these experiments, we examined a potential role for intracellular HMGB1 in modulating phagocytosis. We found that phagocytosis of apoptotic cells resulted in translocation of HMGB1 into the cytoplasm and extracellular space. Transient or stable inhibition of HMGB1 expression in bone marrow-derived macrophages or fibroblasts resulted in increased phagocytosis of apoptotic thymocytes and apoptotic neutrophils. Knockdown of HMGB1 was associated with enhanced activation of Rac-1 and cytoskeletal rearrangement. Intracellular events involved in phagocytosis and upstream of Rac-1 activation, such as phosphorylation of ERK and focal adhesion kinase (FAK), were increased after knockdown of HMGB1. Inhibition of Src kinase activity prevented the increase in phosphorylation of FAK and ERK present during phagocytosis in HMGB1 knockdown cells, and also abrogated the enhancement in phagocytosis associated with HMGB1 knockdown. Interaction between Src and FAK in the cytoplasm of HMGB1 knockdown fibroblasts was enhanced compared with that present in control fibroblasts. Under in vitro conditions, the presence of HMGB1 diminished interactions between purified FAK and Src. These studies demonstrate a novel role for HMGB1 in the regulation of phagocytosis. In particular, these experiments show that intracellular HMGB1, through associating with Src kinase and inhibiting interactions between Src and FAK, diminishes the phagocytic ability of macrophages and other cell populations.


Asunto(s)
Regulación hacia Abajo/inmunología , Proteína HMGB1/fisiología , Líquido Intracelular/inmunología , Líquido Intracelular/metabolismo , Animales , Apoptosis/genética , Apoptosis/inmunología , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Línea Celular , Citofagocitosis/genética , Citofagocitosis/inmunología , Regulación hacia Abajo/genética , Fibroblastos/citología , Fibroblastos/inmunología , Fibroblastos/metabolismo , Técnicas de Silenciamiento del Gen , Células HEK293 , Proteína HMGB1/biosíntesis , Proteína HMGB1/deficiencia , Humanos , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH
15.
J Immunol ; 187(1): 118-25, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21642542

RESUMEN

Although high mobility group box 1 (HMGB1) in tumor cells is involved in many aspects of tumor progression, its role in tumor immune suppression remains elusive. Host cell-derived IL-10 suppressed a naturally acquired CD8 T cell-dependent antitumor response. The suppressive activity of tumor-associated Foxp3(+)CD4(+)CD25(+) regulatory T cells (Treg) was IL-10 dependent. Neutralizing HMGB1 impaired tumor cell-promoted IL-10 production by Treg. Short hairpin RNA-mediated knockdown of HMGB1 (HMGB1 KD) in tumor cells did not affect tumor cell growth but uncovered naturally acquired long-lasting tumor-specific IFN-γ- or TNF-α-producing CD8 T cell responses and attenuated their ability to induce Treg, leading to naturally acquired CD8 T cell- or IFN-γ-dependent tumor rejection. The data suggest that tumor cell-derived HMGB1 may suppress naturally acquired CD8 T cell-dependent antitumor immunity via enhancing Treg to produce IL-10, which is necessary for Treg-mediated immune suppression.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Diferenciación Celular/inmunología , Proteína HMGB1/deficiencia , Proteína HMGB1/genética , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología , Escape del Tumor/inmunología , Animales , Antígenos de Neoplasias/biosíntesis , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/fisiología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/genética , Línea Celular Tumoral , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Neoplasias del Colon/prevención & control , Femenino , Técnicas de Silenciamiento del Gen , Proteína HMGB1/fisiología , Humanos , Inmunidad Innata/genética , Interleucina-10/biosíntesis , Interleucina-10/genética , Interleucina-10/fisiología , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/patología , Neoplasias Mamarias Experimentales/prevención & control , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Linfocitos T Reguladores/metabolismo , Escape del Tumor/genética
16.
J Immunol ; 186(6): 3517-26, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21300822

RESUMEN

Dysregulation of apoptosis is associated with the development of human cancer and resistance to anticancer therapy. We have previously shown in tumor xenografts that DNA alkylating agents induce sporadic cell necrosis and regression of apoptosis-deficient tumors. Sporadic tumor cell necrosis is associated with extracellular release of cellular content such as the high mobility group box 1 (HMGB1) protein and subsequent recruitment of innate immune cells into the tumor tissue. It remained unclear whether HMGB1 and the activation of innate immunity played a role in tumor response to chemotherapy. In this study, we show that whereas DNA alkylating therapy leads to a complete tumor regression in an athymic mouse tumor xenograft model, it fails to do so in tumors deficient in HMGB1. The HMGB1-deficient tumors have an impaired ability to recruit innate immune cells including macrophages, neutrophils, and NK cells into the treated tumor tissue. Cytokine array analysis reveals that whereas DNA alkylating treatment leads to suppression of protumor cytokines such as IL-4, IL-10, and IL-13, loss of HMGB1 leads to elevated levels of these cytokines upon treatment. Suppression of innate immunity and HMGB1 using depleting Abs leads to a failure in tumor regression. Taken together, these results indicate that HMGB1 plays an essential role in activation of innate immunity and tumor clearance in response to DNA alkylating agents.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Resistencia a Antineoplásicos/inmunología , Proteína HMGB1/fisiología , Inmunidad Innata/inmunología , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/prevención & control , Animales , Antineoplásicos Alquilantes/uso terapéutico , Línea Celular Transformada , Línea Celular Tumoral , Células Cultivadas , Células HEK293 , Proteína HMGB1/deficiencia , Proteína HMGB1/metabolismo , Humanos , Masculino , Ratones , Ratones Desnudos , Necrosis , Neoplasias Experimentales/patología , Distribución Aleatoria
17.
Lab Invest ; 90(6): 853-66, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20231821

RESUMEN

Granulomatous nephritis can be triggered by diverse factors and results in kidney failure. However, despite accumulating data about granulomatous inflammation, pathogenetic mechanisms in nephritis remain unclear. The DNA-binding high-mobility group box-1 protein (HMGB1) initiates and propagates inflammation when released by activated macrophages, and functions as an 'alarm cytokine' signaling tissue damage. In this study, we showed elevated HMGB1 expression in renal granulomas in rats with crystal-induced granulomatous nephritis caused by feeding an adenine-rich diet. HMGB1 levels were also raised in urine and serum, as well as in monocyte chemoattractant protein-1 (MCP-1), a mediator of granulomatous inflammation. Injection of HMGB1 worsened renal function and upregulated MCP-1 in rats with crystal-induced granulomatous nephritis. HMGB1 also induced MCP-1 secretion through mitogen-activated protein kinase (MAPK) and phosphoinositide-3-kinase (PI3K) pathways in rat renal tubular epithelial cells in vitro. Hmgb1(+/-) mice with crystal-induced nephritis displayed reduced MCP-1 expression in the kidneys and in urine and the number of macrophages in the kidneys was significantly decreased. We conclude that HMGB1 is a new mediator involved in crystal-induced nephritis that amplifies granulomatous inflammation in a cycle where MCP-1 attracts activated macrophages, resulting in excessive and sustained HMGB1 release. HMGB1 could be a novel target for inhibiting chronic granulomatous diseases.


Asunto(s)
Adenina/farmacología , Granuloma/inducido químicamente , Proteína HMGB1/genética , Enfermedades Renales/inducido químicamente , Nefritis/inducido químicamente , Animales , Nitrógeno de la Urea Sanguínea , Quimiocina CCL2/efectos de los fármacos , Quimiocina CCL2/genética , Creatinina/sangre , Células Epiteliales/citología , Células Epiteliales/fisiología , Granuloma/genética , Granuloma/patología , Granuloma/prevención & control , Proteína HMGB1/deficiencia , Proteína HMGB1/metabolismo , Proteína HMGB1/farmacología , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/patología , Inflamación/prevención & control , Enfermedades Renales/patología , Túbulos Renales/citología , Túbulos Renales/fisiología , Ratones , Nefritis/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Ratas
18.
Nature ; 462(7269): 99-103, 2009 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-19890330

RESUMEN

The activation of innate immune responses by nucleic acids is crucial to protective and pathological immunities and is mediated by the transmembrane Toll-like receptors (TLRs) and cytosolic receptors. However, it remains unknown whether a mechanism exists that integrates these nucleic-acid-sensing systems. Here we show that high-mobility group box (HMGB) proteins 1, 2 and 3 function as universal sentinels for nucleic acids. HMGBs bind to all immunogenic nucleic acids examined with a correlation between affinity and immunogenic potential. Hmgb1(-/-) and Hmgb2(-/-) mouse cells are defective in type-I interferon and inflammatory cytokine induction by DNA or RNA targeted to activate the cytosolic nucleic-acid-sensing receptors; cells in which the expression of all three HMGBs is suppressed show a more profound defect, accompanied by impaired activation of the transcription factors interferon regulatory factor 3 (IRF3) and nuclear factor (NF)-kappaB. The absence of HMGBs also severely impairs the activation of TLR3, TLR7 and TLR9 by their cognate nucleic acids. Our results therefore indicate a hierarchy in the nucleic-acid-mediated activation of immune responses, wherein the selective activation of nucleic-acid-sensing receptors is contingent on the more promiscuous sensing of nucleic acids by HMGBs. These findings may have implications for understanding the evolution of the innate immune system and for the treatment of immunological disorders.


Asunto(s)
Proteínas HMGB/inmunología , Proteínas HMGB/metabolismo , Inmunidad Innata/inmunología , Ácidos Nucleicos/inmunología , Animales , Línea Celular , Citosol/inmunología , ADN/inmunología , Proteínas HMGB/deficiencia , Proteínas HMGB/genética , Proteína HMGB1/deficiencia , Proteína HMGB1/genética , Proteína HMGB1/inmunología , Proteína HMGB1/metabolismo , Proteína HMGB2/deficiencia , Proteína HMGB2/genética , Proteína HMGB2/inmunología , Proteína HMGB2/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Ratones , Ratones Endogámicos C57BL , Modelos Inmunológicos , FN-kappa B/metabolismo , Nucleótidos/química , Nucleótidos/inmunología , Nucleótidos/metabolismo , ARN/inmunología , Transducción de Señal , Receptores Toll-Like/inmunología , Virosis/inmunología , Virosis/virología
19.
Mol Cell Biol ; 27(16): 5650-63, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17548469

RESUMEN

High mobility group box 1 protein (HMGB1) is a chromatin protein that has a dual function as a nuclear factor and as an extracellular factor. Extracellular HMGB1 released by damaged cells acts as a chemoattractant, as well as a proinflammatory cytokine, suggesting that HMGB1 is tightly connected to the process of tissue organization. However, the role of HMGB1 in bone and cartilage that undergo remodeling during embryogenesis, tissue repair, and disease is largely unknown. We show here that the stage-specific secretion of HMGB1 in cartilage regulates endochondral ossification. We analyzed the skeletal development of Hmgb1(-/-) mice during embryogenesis and found that endochondral ossification is significantly impaired due to the delay of cartilage invasion by osteoclasts, osteoblasts, and blood vessels. Immunohistochemical analysis revealed that HMGB1 protein accumulated in the cytosol of hypertrophic chondrocytes at growth plates, and its extracellular release from the chondrocytes was verified by organ culture. Furthermore, we demonstrated that the chondrocyte-secreted HMGB1 functions as a chemoattractant for osteoclasts and osteoblasts, as well as for endothelial cells, further supporting the conclusion that Hmgb1(-/-) mice are defective in cell invasion. Collectively, these findings suggest that HMGB1 released from differentiating chondrocytes acts, at least in part, as a regulator of endochondral ossification during osteogenesis.


Asunto(s)
Cartílago/metabolismo , Proteína HMGB1/metabolismo , Osteogénesis , Animales , Biomarcadores , Huesos/anomalías , Huesos/irrigación sanguínea , Huesos/embriología , Cartílago/citología , Diferenciación Celular , Células Cultivadas , Factores Quimiotácticos/metabolismo , Condrocitos/citología , Condrocitos/metabolismo , Embrión de Mamíferos/anomalías , Placa de Crecimiento/citología , Placa de Crecimiento/metabolismo , Proteína HMGB1/deficiencia , Humanos , Ratones , Modelos Biológicos , Osteoblastos/citología , Osteoblastos/metabolismo , Osteoclastos/citología , Osteoclastos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...