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










Intervalo de ano de publicação
1.
Physiol Behav ; 274: 114418, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38042454

RESUMO

Previous studies have shown the relevance of high mobility group box 1 protein (HMGB1) and tumor necrosis factor α (TNFα) in nerve or tissue injury-induced nociception. However, the role of these proteins in chronic stress and social transfer of stress (STS)-induced dysfunctional pain is not entirely known. The aim of this study was to determine the participation of the spinal HMGB1-TNFα signaling pathway and TNFα receptor 1 (TNFR1) in rats subjected to chronic restraint stress (CRS) and STS. Non-stressed female and male rats in contact with CRS rats increased sniffing behavior of the anogenital area, behavior related to STS. Rats subjected to CRS and STS reduced 50 % withdrawal threshold and reached the value of tactile allodynia after 21 days of stress. Rats return to the basal withdrawal threshold after 30 days without stress and return to allodynia values in only 5 days of stress sessions (priming). Female and male rats subjected to 28 days of CRS or STS were intrathecal injected with glycyrrhizin (inhibitor of HMGB1), thalidomide (inhibitor of the TNFα synthesis), and R7050 (TNFR1 antagonist), in all the cases, an antiallodynic effect was observed. Rats under CRS or STS enhanced HMGB1 and TNFR1 protein expression in DRG and dorsal spinal cord. Data suggest that the spinal HMGB1/TNFα/TNFR1 signaling pathway plays a relevant role in the maintenance of CRS and STS-induced nociceptive hypersensitivity in rats. These proteins could be helpful in developing pain treatments for fibromyalgia in humans.


Assuntos
Proteína HMGB1 , Hiperalgesia , Humanos , Ratos , Masculino , Feminino , Animais , Receptores Tipo I de Fatores de Necrose Tumoral/efeitos adversos , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteína HMGB1/efeitos adversos , Proteína HMGB1/metabolismo , Dor/induzido quimicamente
2.
J Psychiatr Res ; 164: 150-161, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37352811

RESUMO

Morphine is one of the most effective and widely used analgesic drugs. However, chronic morphine use caused opioid-induced hyperalgesia (OIH). The development of OIH limits the use of morphine. The mechanisms of OIH are not fully understood. Toll-like receptor4 (TLR4) and glutamate receptors in the periaqueductal gray (PAG) are critical in OIH, however, the association between TLR4 and N-methyl-D-aspartate Receptors (NMDARs) activation in PAG remains unclear. Microglia activation, increased TLR4/p65 nuclear factor-kappa B (p65 NF-κB) and proinflammatory cytokines in microglia, and phosphorylation of NMDAR1 subunit (NR1) and NMDAR2B subunit (NR2B) in neurons were observed in PAG of OIH mice. Up-regulations of TLR4/p65 NF-κB and proinflammatory cytokines (IL-1ß, IL-6, TNF-α) in BV2 cells were prevented by inhibiting and knocking down TLR4. By inhibiting myeloid differentiation factor 2 (MD2) and knocking down the High-mobility group box 1 (HMGB1), we found that morphine activated TLR4 by HMGB1 but not MD2. We co-cultured Neuro-2a (N2A) with BV2 microglial cell line and found that instead of directly phosphorylating NMDAR subunits, morphine increased the phosphorylation of NR1 and NR2B by inducing TLR4-mediated microglia inflammation. Knocking TLR4 out of PAG by Lentivirus-GFP-TLR4 shRNA reversed these changes and relieved OIH. Our findings suggested that the secretion of HMGB1 induced by morphine-activated TLR4 in microglia, and the proinflammatory factors released by activated microglia phosphorylated NR1 and NR2B of adjacent neurons, induced increased neuronal excitability. In conclusion, TLR4/NMDARs in PAG were involved in the development and maintenance of OIH and supported novel strategies for OIH treatment.


Assuntos
Proteína HMGB1 , Morfina , Camundongos , Animais , Morfina/efeitos adversos , Morfina/metabolismo , Hiperalgesia/induzido quimicamente , Hiperalgesia/metabolismo , NF-kappa B/metabolismo , Microglia/metabolismo , Receptor 4 Toll-Like/metabolismo , Substância Cinzenta Periaquedutal/metabolismo , Transdução de Sinais , Proteína HMGB1/efeitos adversos , Proteína HMGB1/metabolismo , Inflamação/induzido quimicamente , Inflamação/metabolismo , Analgésicos Opioides/efeitos adversos , Citocinas/metabolismo , Neurônios
3.
Immun Inflamm Dis ; 10(11): e711, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36301039

RESUMO

Chronic obstructive pulmonary disease (COPD) is a common chronic respiratory disease with irreversible and continuous progression. It has become the fifth most burdensome disease and the third most deadly disease globally. Therefore, the prevention and treatment of COPD are urgent, and it is also important to clarify the pathogenesis of it. Smoking is the main and most common risk factor for COPD. Cigarette smoke (CS) can cause lung inflammation and other pathological mechanisms in the airways and lung tissue. Airway inflammation is one of the important mechanisms leading to the pathogenesis of COPD. Recent studies have shown that high mobility group box 1 (HMGB1) is involved in the occurrence and development of respiratory diseases, including COPD. HMGB1 is a typical damage-associated molecular pattern (DAMP) protein, which mainly exerts its activity by binding to the receptor for advanced glycation end products (RAGE) and toll-like receptor 4 (TLR4) and further participate in the process of airway inflammation. Studies have shown that the abnormal expression of HMGB1, RAGE, and TLR4 are related to inflammation in COPD. Herein, we discuss the roles of HMGB1, RAGE, and TLR4 in CS/cigarette smoke extract-induced inflammation in COPD, providing a new target for the diagnosis, treatment and prevention of COPD.


Assuntos
Fumar Cigarros , Proteína HMGB1 , Doença Pulmonar Obstrutiva Crônica , Humanos , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Proteína HMGB1/efeitos adversos , Proteína HMGB1/metabolismo , Receptor 4 Toll-Like/metabolismo , Fumar Cigarros/efeitos adversos , Doença Pulmonar Obstrutiva Crônica/induzido quimicamente , Doença Pulmonar Obstrutiva Crônica/metabolismo , Inflamação , Transdução de Sinais
4.
J Bone Miner Res ; 37(9): 1775-1786, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35711109

RESUMO

Medication-related osteonecrosis of the jaw (MRONJ) is a severe complication of antiresorptive or antiangiogenic medications, used in the treatment of bone malignancy or osteoporosis. Bone necrosis, mainly represented by osteocytic death, is always present in MRONJ sites; however, the role of osteocyte death in MRONJ pathogenesis is unknown. High mobility group box 1 (HMGB1) is a non-histone nucleoprotein that in its acetylated form accumulates in the cytoplasm, whereas non-acetylated HMGB1 localizes in the nucleus. SIRT1 deacetylase regulates cellular localization of HMGB1. Interestingly, HMGB1 is released during cell necrosis and promotes inflammation through signaling cascades, including activation of the RAGE receptor. Here, we utilized a well-established mouse MRONJ model that utilizes ligature-induced experimental periodontitis (EP) and treatment with either vehicle or zolendronic acid (ZA). Initially, we evaluated HMGB1-SIRT1 expression in osteocytes at 1, 2, and 4 weeks of treatment. Significantly increased cytoplasmic and perilacunar HMGB1 expression was observed at EP sites of ZA versus vehicle (Veh) animals at all time points. SIRT1 colocalized with cytoplasmic HMGB1 and presented a statistically significant increased expression at the EP sites of ZA animals for all time points. RAGE expression was significantly higher in the submucosal tissues EP sites of ZA animals compared with those in vehicle group. To explore the significance of increased cytoplasmic and extracellular HMGB1 and increased RAGE expression in MRONJ pathogenesis, we used pharmacologic inhibitors of these molecules. Combined HMGB1/RAGE inhibition resulted in lower MRONJ incidence with statistically significant decrease in osteonecrotic areas and bone exposure versus non-inhibitor treated ZA animals. Together, our data point to the role of HMGB1 as a central alarmin, overexpressed at early phase of MRONJ pathogenesis during osteocytic death. Moreover, HMGB1-RAGE pathway may represent a new promising therapeutic target in patients at high risk of MRONJ. © 2022 American Society for Bone and Mineral Research (ASBMR).


Assuntos
Osteonecrose da Arcada Osseodentária Associada a Difosfonatos , Conservadores da Densidade Óssea , Proteína HMGB1 , Osteonecrose , Osteoporose , Periodontite , Animais , Osteonecrose da Arcada Osseodentária Associada a Difosfonatos/tratamento farmacológico , Osteonecrose da Arcada Osseodentária Associada a Difosfonatos/epidemiologia , Conservadores da Densidade Óssea/efeitos adversos , Difosfonatos/efeitos adversos , Proteína HMGB1/efeitos adversos , Proteína HMGB1/metabolismo , Incidência , Camundongos , Osteonecrose/induzido quimicamente , Osteonecrose/tratamento farmacológico , Osteoporose/induzido quimicamente , Sirtuína 1
5.
J Gerontol A Biol Sci Med Sci ; 77(3): 433-442, 2022 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-34723336

RESUMO

The role of cellular senescence in the development of asthma is not well known. We aimed to evaluate the susceptibility of mice with cellular senescence to asthma development and determine whether the mTOR pathway played an important role in this process. Cellular senescence was induced in mice by intranasal instillation of 2% cigarette smoke extract (CSE). Subsequently, a low dose (0.1 µg) of house dust mite (HDM) allergens, which cause no inflammation and airway hyperresponsiveness (AHR) in mice without cellular senescence, was administered intranasally. To evaluate the role of the mTOR pathway in this model, rapamycin (TORC1 inhibitor) was injected intraperitoneally before CSE instillation. CSE significantly increased senescence-associated ß-gal activity in lung homogenate and S100A8/9+ p-mTOR+ population in lung cells. Moreover, S100A8/9+ or HMGB1+ populations in airway epithelial cells with p-mTOR activity increased remarkably. Rapamycin attenuated all changes. Subsequent administration of low-dose HDM allergen induced murine asthma characterized by increased AHR, serum HDM-specific immunoglobulin E, and eosinophilic airway inflammation; these asthma characteristics disappeared after rapamycin injection. In vitro experiments showed significant activation of bone marrow-derived cells cocultured with S100A9 or HMGB1 overexpressing MLE-12 cells treated with HDM allergen, compared to those treated with HDM allergen only. CSE increased the levels of senescence markers (S100A8/9 and HMGB1) in airway epithelial cells, making the mice susceptible to asthma development due to low-dose HDM allergens by activating dendritic cells. Because rapamycin significantly attenuated asthma characteristics, the mTOR pathway may be important in this murine model.


Assuntos
Asma , Fumar Cigarros , Proteína HMGB1 , Alérgenos/efeitos adversos , Alérgenos/metabolismo , Animais , Asma/etiologia , Senescência Celular , Modelos Animais de Doenças , Proteína HMGB1/efeitos adversos , Proteína HMGB1/metabolismo , Pulmão , Camundongos , Camundongos Endogâmicos BALB C , Pyroglyphidae , Sirolimo/metabolismo , Serina-Treonina Quinases TOR/metabolismo
6.
Braz. J. Pharm. Sci. (Online) ; 58: e19791, 2022. tab, graf
Artigo em Inglês | LILACS | ID: biblio-1383988

RESUMO

Abstract In China, Scutellaria is used for treating inflammatory-related diseases. Baicalin is the main active component of Scutellaria and has protective effects on acute pancreatitis. However, the mechanism of Baicalin is still unclear. In this study, the protective effects of baicalin on acute pancreatitis induced by taurocholate and its mechanism are investigated. In this study, mice were randomly divided into three groups: sham operation, model, and treatment groups. Acute pancreatitis in mice was induced by intraperitoneal injection of taurocholate (35 mg/kg). The treatment group was given baicalin (100 mg/kg) 2 h before acute pancreatitis induction. The mRNA expression levels of miR-429, nuclear factor kappa B65(NF-kB65), toll-like receptor 4(TLR4), TNF receptor associated factor6 (TRAF6), NF-kappa-B inhibitor(IkB), Follistatin-like 1 (FSTL1), and interleukin-1 receptor-associated kinase (IRAK) in the liver tissues 24 h after intraperitoneal injection were detected by RT-PCR. Then, the expression levels of NF-kB65, p-NF-κB65, TLR4, TRAF6, IkB, FSTL1, IRAK, p- IRAK, and p- IkB-а proteins were detected by Western blot. IL-6, TNF-α and IL-1 ß in plasma were measured by ELISA, and histopathological changes in the pancreases of the mice were observed. The results showed that after baicalin treatment, miR-429 expression in the pancreatic tissues and the expression levels of NF-kB65, TLR4, TRAF6, p-IkB-а, FSTL1, and p-IRAK decreased. Similarly, pancreatic myeloperoxidase (MPO) activity and the plasma levels of IL-6, TNF-а, IL-12, IL-1ß1, endotoxin, serum amylase, and lipase were reduced. Thus, the pancreatic injury induced by taurocholate was alleviated. The present study indicates that pretreatment with Baicalin can alleviate acute pancreatic injury induced by taurocholate in mice. The mechanism may be associated with the decreased miR-429 expression, reduced FSTL1 signaling pathway activity, TLR4 and TLR4/MyD88 signaling pathway inhibition, and reduced pancreatic inflammation. FSTL1 is the regulatory target for miR-429


Assuntos
Animais , Masculino , Camundongos , Proteína HMGB1/efeitos adversos , Scutellaria/efeitos adversos , Injeções/classificação , Pancreatite/patologia , Ensaio de Imunoadsorção Enzimática/instrumentação , Western Blotting , Receptores do Fator de Necrose Tumoral , Folistatina/administração & dosagem , Fígado/anormalidades
7.
Mol Med Rep ; 23(3)2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33495819

RESUMO

High­mobility group box 1 (HMGB1) is released by necrotic cells and serves an important role in cardiovascular pathology. However, the effects of HMGB1 in cardiomyocyte hypertrophy remain unclear. Therefore, the aim of the present study was to investigate the potential role of HMGB1 in cardiomyocyte hypertrophy and the underlying mechanisms of its action. Neonatal mouse cardiomyocytes (NMCs) were co­cultured with recombinant HMGB1 (rHMGB1). Wortmannin was used to inhibit PI3K activity in cardiomyocytes. Subsequently, atrial natriuretic peptide (ANP), 14­3­3 and phosphorylated­Akt (p­Akt) protein levels were detected using western blot analysis. In addition, nuclear factor of activated T cells 3 (NFAT3) protein levels were measured by western blot analysis and observed in NMCs under a confocal microscope. The results revealed that rHMGB1 increased ANP and p­Akt, and decreased 14­3­3η protein levels. Furthermore, wortmannin abrogated the effects of rHMGB1 on ANP, 14­3­3η and p­Akt protein levels. In addition, rHMGB1 induced nuclear translocation of NFAT3, which was also inhibited by wortmannin pretreatment. The results of this study suggest that rHMGB1 induces cardiac hypertrophy by regulating the 14­3­3η/PI3K/Akt/NFAT3 signaling pathway.


Assuntos
Proteínas 14-3-3/metabolismo , Cardiomegalia/metabolismo , Proteína HMGB1/efeitos adversos , Miócitos Cardíacos/metabolismo , Fatores de Transcrição NFATC/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Cardiomegalia/induzido quimicamente , Cardiomegalia/patologia , Feminino , Proteína HMGB1/farmacologia , Camundongos , Miócitos Cardíacos/patologia , Fatores de Transcrição NFATC/genética , Proteínas Recombinantes
8.
Am J Chin Med ; 44(6): 1145-1166, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27627916

RESUMO

The nucleosomal protein high-mobility group box-1 (HMGB1), which has recently been established as a late mediator of lethal systemic inflammation, has a relatively wide therapeutic window for pharmacological interventions. Compounds produced by marine-derived microbes have been widely investigated for their potential use as bioactive natural products. Cyclic dipeptides, which are also known as diketopiperazines, are molecules that are frequently found in marine-derived microorganisms. While their pharmacological potential has been well established, their biological activities against septic responses have not yet been reported. Here, three diketopiperazines (1-3) isolated from two strains of marine-derived bacteria were investigated for their potential activities against HMGB1-mediated septic responses. The data showed that 1-3 effectively inhibited the lipopolysaccharide (LPS)-induced release of HMGB1 and suppressed the HMGB1-mediated septic responses, including hyperpermeability, leukocyte adhesion and migration, and cell adhesion molecule expression. In addition, 1-3 inhibited the HMGB1-mediated production of tumor necrosis factor-[Formula: see text] (TNF-[Formula: see text] and interleukin (IL)-6 and the activation of nuclear factor-[Formula: see text]B (NF-[Formula: see text]B) and extracellular signal-regulated kinase (ERK) 1 and ERK2. Collectively, these results indicated that 1-3 might act as potential therapeutic agents for various severe vascular inflammatory diseases through the inhibition of the HMGB1 signaling pathway.


Assuntos
Actinomycetales/química , Anti-Infecciosos Locais/farmacologia , Bacillus/química , Dicetopiperazinas/farmacologia , Proteína HMGB1/efeitos adversos , Poríferos/microbiologia , Sepse/tratamento farmacológico , Actinomycetales/isolamento & purificação , Animais , Anti-Infecciosos Locais/química , Anti-Infecciosos Locais/isolamento & purificação , Anti-Infecciosos Locais/uso terapêutico , Bacillus/isolamento & purificação , Sobrevivência Celular/efeitos dos fármacos , Dicetopiperazinas/química , Dicetopiperazinas/isolamento & purificação , Dicetopiperazinas/uso terapêutico , Modelos Animais de Doenças , Sedimentos Geológicos/microbiologia , Proteína HMGB1/fisiologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Camundongos Endogâmicos C57BL , Sepse/genética , Choque Séptico/tratamento farmacológico , Choque Séptico/genética
9.
Yonsei Med J ; 57(5): 1095-105, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27401639

RESUMO

PURPOSE: Appropriate animal models of atherosclerotic plaque are crucial to investigating the pathophysiology of atherosclerosis, as well as for the evaluation of the efficacy and safety of vascular devices. We aimed to develop a novel animal model that would be suitable for the study of advanced atherosclerotic lesions in vivo. MATERIALS AND METHODS: Atherosclerotic plaque was induced in 24 iliac arteries from 12 rabbits by combining a high cholesterol diet, endothelial denudation, and injection into the vessel wall with either saline (n=5), olive oil (n=6), or inflammatory proteins [n=13, high-mobility group protein B1 (HMGB1) n=8 and tumor necrosis factor (TNF)-α n=5] using a Cricket™ Micro-infusion catheter. Optical coherence tomography (OCT) was performed to detect plaque characteristics after 4 weeks, and all tissues were harvested for histological evaluation. RESULTS: Advanced plaque was more frequently observed in the group injected with inflammatory proteins. Macrophage infiltration was present to a higher degree in the HMGB1 and TNF-α groups, compared to the oil or saline group (82.1±5.1% and 94.6±2.2% compared to 49.6±14.0% and 46.5±9.6%, p-value<0.001), using RAM11 antibody staining. On OCT, lipid rich plaques were more frequently detected in the inflammatory protein group [saline group: 2/5 (40%), oil group: 3/5 (50%), HMGB1 group: 6/8 (75%), and TNF-α group: 5/5 (100%)]. CONCLUSION: These data indicate that this rabbit model of atherosclerotic lesion formation via direct injection of pro-inflammatory proteins into the vessel wall is useful for in vivo studies investigating atherosclerosis.


Assuntos
Modelos Animais de Doenças , Proteína HMGB1/efeitos adversos , Placa Aterosclerótica/induzido quimicamente , Fator de Necrose Tumoral alfa/efeitos adversos , Animais , Colesterol na Dieta/administração & dosagem , Endotélio/cirurgia , Artéria Ilíaca/diagnóstico por imagem , Artéria Ilíaca/patologia , Artéria Ilíaca/cirurgia , Injeções Intra-Arteriais , Macrófagos , Masculino , Azeite de Oliva/efeitos adversos , Placa Aterosclerótica/diagnóstico por imagem , Placa Aterosclerótica/patologia , Coelhos , Cloreto de Sódio/efeitos adversos , Tomografia de Coerência Óptica
10.
Yonsei Medical Journal ; : 1095-1105, 2016.
Artigo em Inglês | WPRIM (Pacífico Ocidental) | ID: wpr-34056

RESUMO

PURPOSE: Appropriate animal models of atherosclerotic plaque are crucial to investigating the pathophysiology of atherosclerosis, as well as for the evaluation of the efficacy and safety of vascular devices. We aimed to develop a novel animal model that would be suitable for the study of advanced atherosclerotic lesions in vivo. MATERIALS AND METHODS: Atherosclerotic plaque was induced in 24 iliac arteries from 12 rabbits by combining a high cholesterol diet, endothelial denudation, and injection into the vessel wall with either saline (n=5), olive oil (n=6), or inflammatory proteins [n=13, high-mobility group protein B1 (HMGB1) n=8 and tumor necrosis factor (TNF)-α n=5] using a Cricket™ Micro-infusion catheter. Optical coherence tomography (OCT) was performed to detect plaque characteristics after 4 weeks, and all tissues were harvested for histological evaluation. RESULTS: Advanced plaque was more frequently observed in the group injected with inflammatory proteins. Macrophage infiltration was present to a higher degree in the HMGB1 and TNF-α groups, compared to the oil or saline group (82.1±5.1% and 94.6±2.2% compared to 49.6±14.0% and 46.5±9.6%, p-value<0.001), using RAM11 antibody staining. On OCT, lipid rich plaques were more frequently detected in the inflammatory protein group [saline group: 2/5 (40%), oil group: 3/5 (50%), HMGB1 group: 6/8 (75%), and TNF-α group: 5/5 (100%)]. CONCLUSION: These data indicate that this rabbit model of atherosclerotic lesion formation via direct injection of pro-inflammatory proteins into the vessel wall is useful for in vivo studies investigating atherosclerosis.


Assuntos
Animais , Masculino , Coelhos , Colesterol na Dieta/administração & dosagem , Modelos Animais de Doenças , Endotélio/cirurgia , Proteína HMGB1/efeitos adversos , Artéria Ilíaca/diagnóstico por imagem , Injeções Intra-Arteriais , Macrófagos , Azeite de Oliva/efeitos adversos , Placa Aterosclerótica/induzido quimicamente , Cloreto de Sódio/efeitos adversos , Tomografia de Coerência Óptica , Fator de Necrose Tumoral alfa/efeitos adversos
11.
Am J Chin Med ; 43(5): 991-1012, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26224030

RESUMO

The ubiquitous nuclear protein, high mobility group box 1 (HMGB1), is released by activated macrophages and human umbilical vein endothelial cells (HUVECs) and functions as a late mediator of experimental sepsis. Aspalathin (Asp) and nothofagin (Not), which have been reported to have anti-oxidant activity, are the two major active dihydrochalcones found in green rooibos. In this study, we investigated the antiseptic effects and underlying mechanisms of Asp and Not against HMGB1-mediated septic responses in HUVECs and mice. The anti-inflammatory activities of Asp and Not were determined by measuring permeability, monocyte adhesion and migration, and activation of proinflammatory proteins in HMGB1-activated HUVECs and mice. According to the results, Asp and Not effectively inhibited lipopolysaccharide (LPS)-induced release of HMGB1, and suppressed HMGB1-mediated septic responses, such as hyperpermeability, adhesion and migration of leukocytes, and expression of cell adhesion molecules. In addition, Asp and Not suppressed the production of tumor necrosis factor alpha (TNF-α) and interleukin 6 (IL-6), the activation of nuclear factor-κB (NF-κB) and extracellular signal-regulated kinases 1 and 2 (ERK1/2) by HMGB1. Collectively, these results indicate that Asp and Not could be potential therapeutic agents for the treatment of various severe vascular inflammatory diseases via the inhibition of the HMGB1 signaling pathway.


Assuntos
Anti-Inflamatórios , Aspalathus/química , Chalconas/farmacologia , Chalconas/uso terapêutico , Proteína HMGB1/efeitos adversos , Proteína HMGB1/fisiologia , Fitoterapia , Sepse/tratamento farmacológico , Sepse/genética , Animais , Antioxidantes , Chalconas/isolamento & purificação , Modelos Animais de Doenças , Células Endoteliais da Veia Umbilical Humana , Humanos , Interleucina-6/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo
12.
Food Chem Toxicol ; 52: 97-104, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23146691

RESUMO

High mobility group box 1 (HMGB1) protein acts as a potent proinflammatory cytokine and is involved in the pathogenesis of several vascular diseases, such as, systemic vasculitis and sepsis. Emodin-6-O-ß-D-glucoside (EG) is a new active compound from Reynoutria japonica, and its biologic activities have not been previously investigated. In this study, we first investigated the antiinflammatory activities of EG on HMGB1-mediated proinflammatory responses in human umbilical vein endothelial cells (HUVECs) and in a murine cecal ligation and puncture (CLP)-model of sepsis in mice. EG was found to suppress the release of HMGB1, the production of tumor necrosis factor (TNF)-α, and the activation of nuclear factor-κB (NF-κB) by HMGB1 in HUVECs, and to inhibit HMGB1-mediated hyperpermeability and leukocyte migration in mice. In the CLP model, HMGB1 was highly released, but this release was prevented by EG. Furthermore, EG also increased the survival times of CLP administered mice. Collectively, this study shows EG can protect barrier integrity and inhibit HMGB1-mediated inflammatory responses, which suggests a potential use as a therapy for sepsis or septic shock.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Emodina/análogos & derivados , Glucosídeos/farmacologia , Proteína HMGB1/efeitos adversos , Proteína HMGB1/metabolismo , Inflamação/tratamento farmacológico , Animais , Ceco/cirurgia , Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Citoesqueleto/efeitos dos fármacos , Modelos Animais de Doenças , Emodina/farmacologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Inflamação/induzido quimicamente , Inflamação/metabolismo , Ligadura , Lipopolissacarídeos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/efeitos dos fármacos , NF-kappa B/metabolismo , Sepse/tratamento farmacológico , Sepse/mortalidade , Receptor 4 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
13.
Pathobiology ; 77(4): 210-7, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20616616

RESUMO

OBJECTIVE: High-mobility group box 1 (HMGB1) was closely associated with progression and metastasis of colorectal cancer. METHODS: We examined the significance of HMGB1 in causing colon carcinogenesis induced by azoxymethane (AOM) injection in Fischer 344 rats fed on a control diet (group C), a 15% linoleic acid (LA) diet (group L), a control diet with 10% glucose drink (group G), and a 15% LA diet with a 10% glucose drink (group L+G). RESULTS: Group L+G showed the highest body weight and calorie intake. Serum and mucosal HMGB1 levels were temporally increased in all groups, while the highest levels were observed in group L+G. Mucosal HMGB1 levels were correlated with cancer multiplicity and nodal metastases. In the AOM-injected rats fed the 15% LA diet with 10% glucose drink, administration of HMGB1 antibody suppressed serum HMGB1 concentration and cancer multiplicity. CONCLUSION: These data suggest that dietary LA and glucose provided the synergistic effect on AOM-induced rat colon cancer through HMGB1 induction.


Assuntos
Azoximetano/toxicidade , Carcinógenos/toxicidade , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/metabolismo , Proteína HMGB1/metabolismo , Ácido Linoleico/efeitos adversos , Animais , Azoximetano/metabolismo , Azoximetano/farmacologia , Neoplasias do Colo/patologia , Dieta , Sinergismo Farmacológico , Glucose/efeitos adversos , Glucose/metabolismo , Glucose/farmacologia , Proteína HMGB1/efeitos adversos , Proteína HMGB1/farmacologia , Ácido Linoleico/administração & dosagem , Ácido Linoleico/metabolismo , Ácido Linoleico/farmacologia , Masculino , Distribuição Aleatória , Ratos , Ratos Endogâmicos F344
14.
Injury ; 41(1): 21-6, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19729158

RESUMO

Despite the enormous efforts to elucidate the mechanisms of the development of multiple organ failure (MOF) following trauma, MOF following trauma is still a leading cause of late post-injury death and morbidity. Now, it has been proven that excessive systemic inflammation following trauma participates in the development of MOF. Fundamentally, the inflammatory response is a host-defence response; however, on occasion, this response turns around to cause deterioration to host depending on exo- and endogenic factors. Through this review we aim to describe the pathophysiological approach for MOF after trauma studied so far and also introduce the prospects of this issue for the future.


Assuntos
Proteínas do Sistema Complemento/fisiologia , Citocinas/imunologia , Insuficiência de Múltiplos Órgãos/fisiopatologia , Espécies Reativas de Oxigênio/efeitos adversos , Traumatismo por Reperfusão/fisiopatologia , Ferimentos e Lesões/complicações , Ativação do Complemento/fisiologia , Proteínas do Sistema Complemento/metabolismo , Citocinas/metabolismo , Progressão da Doença , Proteína HMGB1/efeitos adversos , Proteína HMGB1/fisiologia , Humanos , Mucosa Intestinal/metabolismo , Masculino , Insuficiência de Múltiplos Órgãos/imunologia , Neutrófilos/imunologia , Espécies Reativas de Oxigênio/metabolismo , Síndrome do Desconforto Respiratório/imunologia , Sepse/imunologia , Síndrome de Resposta Inflamatória Sistêmica/imunologia
15.
Int J Pharm ; 375(1-2): 140-7, 2009 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-19442462

RESUMO

The relatively low efficiency of transgene expression is a major drawback of non-viral gene delivery systems despite it being a safer alternative for gene therapy. Modifications of non-viral carriers with peptides have been utilized to improve their gene transfer efficiency. In this study, we described a new combined carrier, which was comprised of a cationic polymer, polyethylenimine (PEI), and a nuclear protein, high mobility group box 1 (HMGB1) containing nuclear localization sequences (NLS). The HMGB1/branched or linear PEI (b-PEI or l-PEI) combined carriers have been investigated for their cytotoxicity and ability to condense and deliver plasmid DNA (pDNA) to mammalian cells in vitro. Both HMGB1 and PEI formed complexes with pDNA are revealed by a gel-retardation assay. Compared with pDNA/l-PEI complexes, a reduction in particle size was observed for pDNA/HMGB1/l-PEI ternary complexes. In MTT assay the results of cell viability suggested lower cytotoxicity for HMGB1/PEI combined carriers. Transfection efficiencies of pDNA/HMGB1/PEI ternary complexes were evaluated by green fluorescent protein expression level measured by fluorescence spectroscopy and flow cytometry. Transfection efficiencies for pDNA/HMGB1/l-PEI ternary complexes and pDNA/HMGB1/b-PEI ternary complexes showed more than 2.9-fold and 4.0-fold greater than that for pDNA/l-PEI complexes and pDNA/b-PEI complexes, respectively. Thus, our results showed that HMGB1/PEI combined carriers may be a versatile non-viral carrier for pDNA with high transfection efficiency.


Assuntos
DNA/administração & dosagem , Terapia Genética/métodos , Proteína HMGB1/química , Polietilenoimina/química , Sobrevivência Celular/efeitos dos fármacos , Citometria de Fluxo , Regulação da Expressão Gênica , Técnicas de Transferência de Genes , Vetores Genéticos/efeitos adversos , Vetores Genéticos/química , Proteínas de Fluorescência Verde/genética , Proteína HMGB1/efeitos adversos , Células HeLa , Humanos , Tamanho da Partícula , Plasmídeos/administração & dosagem , Polietilenoimina/efeitos adversos , Espectrometria de Fluorescência , Transfecção/métodos
16.
J Immunol ; 180(12): 8369-77, 2008 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-18523304

RESUMO

Sepsis, a life-threatening complication of infections and the most common cause of death in intensive care units, is characterized by a hyperactive and out-of-balance network of endogenous proinflammatory cytokines. None of the current therapies are entirely effective, illustrating the need for novel therapeutic approaches. Ghrelin (GHR) is an orexigenic peptide that has emerged as a potential endogenous anti-inflammatory factor. In this study, we show that the delayed administration of GHR protects against the mortality in various models of established endotoxemia and sepsis. The therapeutic effect of GHR is mainly mediated by decreasing the secretion of the high mobility box 1 (HMGB1), a DNA-binding factor that acts as a late inflammatory factor critical for sepsis progression. Macrophages seem to be the major cell targets in the inhibition of HMGB1 secretion, in which GHR blocked its cytoplasmic translocation. Interestingly, we also report that GHR shows a potent antibacterial activity in septic mice and in vitro. Remarkably, GHR also reduces the severity of experimental arthritis and the release of HMGB1 to serum. Therefore, by regulating crucial processes of sepsis, such as the production of early and late inflammatory mediators by macrophages and the microbial load, GHR represents a feasible therapeutic agent for this disease and other inflammatory disorders.


Assuntos
Peptídeos Catiônicos Antimicrobianos/uso terapêutico , Grelina/uso terapêutico , Proteína HMGB1/antagonistas & inibidores , Proteína HMGB1/metabolismo , Sepse/microbiologia , Sepse/prevenção & controle , Animais , Peptídeos Catiônicos Antimicrobianos/administração & dosagem , Peptídeos Catiônicos Antimicrobianos/antagonistas & inibidores , Peptídeos Catiônicos Antimicrobianos/fisiologia , Artrite Experimental/imunologia , Artrite Experimental/metabolismo , Artrite Experimental/terapia , Atividade Bactericida do Sangue/imunologia , Células Cultivadas , Regulação para Baixo/imunologia , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/mortalidade , Infecções por Escherichia coli/prevenção & controle , Grelina/administração & dosagem , Grelina/antagonistas & inibidores , Grelina/fisiologia , Proteína HMGB1/efeitos adversos , Proteína HMGB1/sangue , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos DBA , Transporte Proteico/imunologia , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/efeitos adversos , Proteínas Recombinantes/metabolismo , Sepse/imunologia , Sepse/mortalidade , Fatores de Tempo
18.
Scand J Immunol ; 65(6): 530-7, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17523945

RESUMO

Rheumatoid arthritis (RA) is an autoimmune disease characterized by a persistent inflammation of the synovium, leading to the erosion of articular cartilage and bone. Synovial mast cells and their effector molecule, histamine, receive increased attention as mediators of joint inflammation. The aim of our study was to analyse levels of free histamine in serum and joint fluid of RA patients and to evaluate the potential inflammatogenic properties of histamine in vivo and in vitro. Histamine levels were measured by an ELISA in synovial fluid and sera of RA patients and of healthy controls. Histamine levels were also assessed in plasma of RA patients undergoing anti-TNF-alpha treatment. In the murine part of the study, histamine was injected intra-articularly in the knee joint of mice and the joints were subsequently analysed with respect to induction of inflammation. RA patients displayed significantly lower levels of histamine in circulation (0.93 +/- 0.16 ng/ml) compared with the healthy controls (1.89 +/- 0.45 ng/ml, P < 0.001). Locally, in synovial fluid the levels of histamine were even lower (0.37 +/- 0.16 ng/ml, P < 0.0006). Long-term anti-TNF-alpha treatment significantly increased circulating levels of histamine in RA patients. Our experiments on animals show that histamine on its own neither induces inflammation in the joint cavity nor influences the course of HMGB1 and peptidoglycan-induced joint inflammation. Based on our experimental and clinical studies we suggest that histamine lacks harmful properties in RA.


Assuntos
Artrite Reumatoide/sangue , Artrite Reumatoide/tratamento farmacológico , Agonistas dos Receptores Histamínicos/uso terapêutico , Liberação de Histamina/efeitos dos fármacos , Histamina/sangue , Articulação do Joelho/efeitos dos fármacos , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Artrite Reumatoide/induzido quimicamente , Artrografia , Células Cultivadas , Feminino , Proteína HMGB1/administração & dosagem , Proteína HMGB1/efeitos adversos , Liberação de Histamina/imunologia , Humanos , Injeções Intra-Articulares , Articulações , Articulação do Joelho/ultraestrutura , Masculino , Camundongos , Camundongos Endogâmicos , Pessoa de Meia-Idade , Valores de Referência , Líquido Sinovial/química , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/sangue
19.
Curr Opin Pharmacol ; 6(2): 130-5, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16487750

RESUMO

A nuclear protein, high mobility group box 1 (HMGB1), is released passively by necrotic cells, and actively by macrophages/monocytes in response to exogenous and endogenous inflammatory stimuli. After binding to the receptor for advanced glycation end products (RAGE) or toll-like receptor 4 (TLR4), HMGB1 activates vascular endothelial cells and macrophages/monocytes to express proinflammatory cytokines, chemokines and adhesion molecules. Pharmacological suppression of its activities or release is protective against lethal endotoxemia and sepsis, establishing HMGB1 as a critical mediator of lethal systemic inflammation. In light of the pathogenic role of inflammation in cardiovascular diseases, we propose that HMGB1, a proinflammatory cytokine derived from both injured endothelium and activated macrophages/monocytes, could contribute to the progression of atherosclerosis and other cardiovascular diseases.


Assuntos
Doenças Cardiovasculares/etiologia , Proteína HMGB1 , Inflamação/etiologia , Sequência de Aminoácidos , Proteína HMGB1/efeitos adversos , Proteína HMGB1/metabolismo , Proteína HMGB1/fisiologia , Humanos , Dados de Sequência Molecular , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...