Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Interferon Cytokine Res ; 43(1): 43-52, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36603105

RESUMO

High altitude hypoxia can lead to a spectrum of gastrointestinal problems. As the first line of host immune defense, innate immune response in the intestinal mucosa plays a pivotal role in maintaining intestinal homeostasis and protecting against intestinal injury at high altitude. This study aimed to investigate the effect of hypoxia on the colonic mucosal barrier and toll-like receptor 4 (TLR4)-mediated innate immune responses in the colon. The mice were exposed to a hypobaric chamber to simulate a 5,000 m plateau environment for 7 days, and the colonic mucosa changes were recorded. At the same time, the inflammation model was established by lipopolysaccharide (LPS) to explore the effects of hypoxia on the TLR4/nuclear factor kappa B (NF-κB) signaling pathway and its downstream inflammatory factors [tumor necrosis factor-α, interleukin (IL)-1ß, IL-6, and interferon (IFN)-γ] in the colon. We found that hypoxic exposure caused weight loss and structural disturbance of the colonic mucosa in mice. Compared with the control group, the protein levels of TLR4 [fold change (FC) = 0.75 versus FC = 0.23], MyD88 (FC = 0.80 versus FC = 0.30), TIR-domain-containing adaptor protein inducing interferon-ß (TRIF: FC = 0.89 versus FC = 0.38), and NF-κB p65 (FC = 0.75 versus FC = 0.24) in the colon of mice in the hypobaric hypoxia group were significantly decreased. LPS-induced upregulation of the TLR4/NF-κB signaling and its downstream inflammatory factors was inhibited by hypoxia. Specifically, compared with the LPS group, the protein levels of TLR4 (FC = 1.18, FC = 0.86), MyD88 (FC = 1.20, FC = 0.80), TRIF (FC = 1.20, FC = 0.86), and NF-κB p65 (FC = 1.29, FC = 0.62) and the mRNA levels of IL-1ß (FC = 7.38, FC = 5.06), IL-6 (FC = 16.06, FC = 9.22), and IFN-γ (FC = 2.01, FC = 1.16) were reduced in the hypobaric hypoxia plus LPS group. Our findings imply that hypoxia could lead to marked damage of the colonic mucosa and a reduction of TLR4-mediated colonic innate immune responses, potentially reducing host defense responses to colonic pathogens.


Assuntos
Hipóxia , Imunidade Inata , NF-kappa B , Receptor 4 Toll-Like , Animais , Camundongos , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/farmacologia , Colo/imunologia , Colo/patologia , Hipóxia/imunologia , Hipóxia/metabolismo , Interleucina-6/metabolismo , Lipopolissacarídeos/farmacologia , Fator 88 de Diferenciação Mieloide/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais , Receptor 4 Toll-Like/genética
2.
Mol Biol Rep ; 50(3): 2545-2557, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36611117

RESUMO

BACKGROUND: Neuronal apoptosis is the main cause for the disabilities and deaths of patients suffered with stroke. Neuroprotectants are clinically used to reduce neuronal apoptosis in ischemic stroke. However, the current neuroprotectants have multiple limitations. Myricetin is beneficial for multiple neurodegenerative diseases, but the role of myricetin as a neuroprotective agent in ischemic stroke is still not fully understood. METHODS AND RESULTS: Middle cerebral artery occlusion, Terminal deoxynucleotidyl transferase dUTP nick-end labeling staining and Western blots were used to explore the anti-apoptotic effects of myricetin in vivo. Flow cytometry, Western blots and Ca2+ staining were used to study the neuroprotective effects of myricetin in vitro. In this study, we first demonstrated that myricetin reduced neuronal apoptosis after ischemia in vivo and in vitro. And, among the factors of apoptosis after ischemic stroke, excitotoxicity, oxidative stress and inflammation-induced apoptosis can be alleviated by myricetin. Moreover, we further demonstrated that myricetin was able to improve neuronal intrinsic apoptosis by inhibiting the phosphorylation of extracellular signal-regulated kinase in the oxygen and glucose deprivation in vitro. CONCLUSIONS: Summarily, our results support myricetin as a novel neuroprotectant for the prevention or treatment of ischemic stroke via MAPK-ERK signaling pathway.


Assuntos
Isquemia Encefálica , AVC Isquêmico , Fármacos Neuroprotetores , Traumatismo por Reperfusão , Acidente Vascular Cerebral , Humanos , Sistema de Sinalização das MAP Quinases , Fármacos Neuroprotetores/farmacologia , Acidente Vascular Cerebral/tratamento farmacológico , Apoptose , Isquemia Encefálica/tratamento farmacológico
3.
Gut Microbes ; 13(1): 1972757, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34592891

RESUMO

Hyaluronan is a glycosaminoglycan polymer that has been shown to play an important role in homeostasis of the gastrointestinal tract. However, its mechanistic significance in gastrointestinal epithelial barrier elements remain unexplored. Here, our results revealed that hyaluronan treatment resulted in significant changes in the gut microbiota in mice. To demonstrate the functional consequences of hyaluronan-treatment and hyaluronan-induced microbiota alterations, Citrobacter rodentium- and DSS-induced colitis models and microbiota transplantation approaches were utilized. We showed that hyaluronan alleviated intestinal inflammation in both pathogen and chemically induced intestinal mucosal damage. The protection in bacterial colitis was associated with enhanced C. rodentium clearance and alleviation of pathogen-induced gut dysbiosis. Microbiota transplantation experiments showed that the hyaluronan-altered microbiota is sufficient to confer protection against C. rodentium infection. Colonization with Akkermansia muciniphila, a commensal bacterium that is greatly enriched by hyaluronan treatment, alleviated C. rodentium-induced bacterial colitis in mice. Additionally, Akkermansia-induced protection was found to be associated with the induction of goblet cells and the production of mucins and epithelial antimicrobial peptides. Collectively, these results provide novel insights into the regulatory role of hyaluronan in modulating the gut microbiota and immunity in enteric infection and inflammation, with therapeutic potential for gut microbiome-targeted immunotherapy.


Assuntos
Citrobacter rodentium/fisiologia , Colite/prevenção & controle , Infecções por Enterobacteriaceae/prevenção & controle , Microbioma Gastrointestinal/efeitos dos fármacos , Ácido Hialurônico/administração & dosagem , Mucosa Intestinal/imunologia , Animais , Bactérias/classificação , Bactérias/efeitos dos fármacos , Bactérias/genética , Bactérias/isolamento & purificação , Colite/imunologia , Colite/microbiologia , Infecções por Enterobacteriaceae/imunologia , Infecções por Enterobacteriaceae/microbiologia , Feminino , Humanos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/microbiologia , Camundongos , Camundongos Endogâmicos BALB C
4.
Bioengineered ; 12(1): 7985-7994, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34666625

RESUMO

High altitude hypoxia (HAH) involves the pathogenesis of ulcerative colitis (UC) and gastrointestinal erosions. However, the mechanism of effects of HAH in colitis remains controversial. This study reports the immunomodulation mediated by HAH to enhancing the severity of UC in the mice model. BALB/c mice were used to establish the UC model by dextran sulfate sodium (DSS) compared to wild type mice. Mice groups were exposed to hypoxic conditions in a hypobaric chamber with an altitude of 5000 m for 7 days. Then, Spleen, mesenteric lymph nodes and colon tissues were collected. The activity of UC, the infiltration of the immune cells, and the released cytokines were investigated. Results showed that the severity of DSS-induced UC significantly increased in mice exposed to HAH. The analysis of pathological changes showed increased weight loss and decreased colon length accompanied by diarrhea and bloody feces in the hypobaric hypoxia group. Interestingly, the levels of inflammatory cytokines IL-17, TNF-α, and IFN-γ in the spleen and mesenteric lymph node showed a significant increase within the colon of the hypobaric hypoxia group. The population of Th 1 and Th 17 cells in the spleen was significantly increased in mice exposed to hypobaric hypoxia compared NC group. Suggesting that high altitude hypoxia enhances colitis in mice through activating the increase of inflammatory Th1 and Th17 lymphocytes. In conclusion, this study revealed that hypobaric hypoxia directly increases the severity of UC in the mice model via increasing the activity of inflammatory CD4+ Th1 and Th 17 lymphocytes.


Assuntos
Doença da Altitude/imunologia , Colite/imunologia , Sulfato de Dextrana/efeitos adversos , Linfócitos/metabolismo , Células Th1/imunologia , Células Th17/imunologia , Animais , Colite/induzido quimicamente , Modelos Animais de Doenças , Feminino , Interferon gama/metabolismo , Interleucina-17/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Fator de Necrose Tumoral alfa/metabolismo
5.
Gen Physiol Biophys ; 40(3): 183-195, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-34100375

RESUMO

Hypoxia exposure often cause the increases of pulmonary arterial pressure (PAP). Studies reported that mast cells (MCs) participate in pulmonary vascular remodeling and promote the formation of chronic pulmonary hypertension. Current studies mainly focus on the change of MCs under chronic hypoxia, but few studies on the regulatory role and mechanism of MCs under acute hypoxia. Therefore, present study investigated the dynamic change of MCs in lung tissues under acute hypoxia and the role of MCs activation in the increasement of PAP. In our study, we established an experimental rat model of acute hypobaric hypoxia using a hypobaric chamber (simulated altitude of 7,000 m) and pretreated with MCs degranulation inhibitor sodium cromoglycate (SCG) to study the MCs changes under acute hypoxic exposure. We found that acute hypobaric hypoxia contributed to the increased quantity, activity, and degranulation of MCs and SCG pretreatment showed attenuated PAP elevation under acute hypoxia. Our findings implied that there is a possible mechanism of acute hypoxia cause rapid recruitment of MCs, activation, and explosive degranulation to release Tryptase, Chymase, IL-6, His, 5-HT, and Ang II, which further contributed to pulmonary microvascular contraction and increase in PAP. This work extends the knowledge about MCs, providing a potential profile of MCs as an alternative treatment for high-altitude pulmonary edema (HAPE)-related increased pulmonary artery pressure.


Assuntos
Doença da Altitude , Hipertensão Pulmonar , Animais , Hipóxia , Pulmão , Mastócitos , Artéria Pulmonar , Ratos
6.
Biomed Pharmacother ; 129: 110477, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32768962

RESUMO

OBJECTIVE: The pathogenesis and mechanism of colitis may be related to intestinal flora, genetic susceptibility, environmental and immune factors. Among these various factors, the importance of environmental factors in the pathogenesis of colitis has been increasingly recognized. The purpose of this study was to investigate the effects of hypoxia on intestinal mucosal immunity. METHODS: Experimental colitis was induced by oral gavage of Citrobacter rodentium (C. rodentium) in mice, then divided into normoxia group and hypoxia group. Mice were sacrificed after 2 weeks. Physiological and blood biochemical indicators were monitored to verify the hypoxia model. The body weight, fecal bacterial output, colon length and colon histopathology were observed to evaluate severity of colitis. The concentration of cytokines in colonic tissues were detected by ELISA. The percentage of CD4+ IFN-γ+ (Th1) and CD4+ IL-17+ (Th17) cells in mesenteric lymph nodes (MLN) were detected by flow cytometry. The levels of mucosal antimicrobial peptides (AMPs), related inflammatory factors and transcription factors in colon tissues were detected by qRT-PCR. RESULTS: Mice in hypoxic C. rodentium infection (Hypoxia + C.r.) group exhibited significant decrease in body weight, increase in fecal bacterial pathogen output, and more severe histopathological damage in the colon compared with the C. rodentium infection (Nomoxia + C.r.) group. Meanwhile, the level of NF-κB, TLR4, COX-2, IL-6 and TNF-α of colonic tissue were increased, while IL17, IL-22, and Reg3γ were decreased. The percentage of CD4+ IFN-γ+ (Th1) and CD4+ IL-17+ (Th17) cells in MLN were significantly decreased in mice of Hypoxia + C.r. group, accompanied by the decreased of IFN-γ and IL-17. In addition, the level of the T-bet, RORγt, IL-12 and IL-23 were decreased in mice of Hypoxia + C.r. group. CONCLUSIONS: Hypoxic exposure significantly exacerbates the symptoms and the pathological damage of mice with colitis and influences the immune function by down-regulating Th1 and Th17 responses in C. rodentium-induced colitis in mice.


Assuntos
Citrobacter rodentium/imunologia , Colite/imunologia , Colo/imunologia , Infecções por Enterobacteriaceae/imunologia , Hipóxia/imunologia , Imunidade nas Mucosas , Mucosa Intestinal/imunologia , Animais , Colite/metabolismo , Colite/microbiologia , Colite/patologia , Colo/metabolismo , Colo/microbiologia , Colo/patologia , Citocinas/metabolismo , Modelos Animais de Doenças , Infecções por Enterobacteriaceae/metabolismo , Infecções por Enterobacteriaceae/microbiologia , Infecções por Enterobacteriaceae/patologia , Feminino , Interações Hospedeiro-Patógeno , Hipóxia/patologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Camundongos Endogâmicos BALB C , NF-kappa B/metabolismo , Exacerbação dos Sintomas , Células Th1/imunologia , Células Th1/metabolismo , Células Th1/microbiologia , Células Th17/imunologia , Células Th17/metabolismo , Células Th17/microbiologia , Receptor 4 Toll-Like/metabolismo
7.
Exp Physiol ; 105(9): 1660-1668, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32706493

RESUMO

NEW FINDINGS: What is the central question of this study? The acute hypoxic compensatory reaction is based on haemodynamic changes, and ß-adrenoceptors are involved in haemodynamic regulation. What is the role of ß-adrenoceptors in haemodynamics during hypoxic exposure? What is the main finding and its importance? Activation of ß2 -adrenoceptors attenuates the increase in pulmonary artery pressure during hypoxic exposure. This compensatory reaction activated by ß2 -adrenoceptors during hypoxic stress is very important to maintain the activities of normal life. ABSTRACT: The acute hypoxic compensatory reaction is accompanied by haemodynamic changes. We monitored the haemodynamic changes in rats undergoing acute hypoxic stress and applied antagonists of ß-adrenoceptor (ß-ARs) subtypes to reveal the regulatory role of ß-ARs on haemodynamics. Sprague-Dawley rats were randomly divided into control, atenolol (ß1 -AR antagonist), ICI 118,551 (ß2 -AR antagonist) and propranolol (non-selective ß-AR antagonist) groups. Rats were continuously recorded for changes in haemodynamic indexes for 10 min after administration. Then, a hypoxic ventilation experiment [15% O2 , 2200 m a.sl., 582 mmHg (0.765 Pa), PO2 87.3 mmHg; Xining, China] was conducted, and the indexes were monitored for 5 min after induction of hypoxia. Plasma catecholamine concentrations were also measured. We found that, during normoxia, the mean arterial pressure, heart rate, ascending aortic blood flow and pulmonary artery pressure were reduced in the propranolol and atenolol groups. Catecholamine concentrations were increased significantly in the atenolol group compared with the control group. During hypoxia, mean arterial pressure and total peripheral resistance were decreased in the control, propranolol and ICI 118,551 groups. Pulmonary arterial pressure and pulmonary vascular resistance were increased in the propranolol and ICI 118,551 groups. During hypoxia, catecholamine concentrations were increased significantly in the control group, but decreased in ß-AR antagonist groups. In conclusion, the ß2 -AR is involved in regulation of pulmonary haemodynamics in the acute hypoxic compensatory reaction, and the activation of ß2 -ARs attenuates the increase in pulmonary arterial pressure during hypoxic stress. This compensatory reaction activated by ß2 -ARs during hypoxic stress is very important to maintain activities of normal life.


Assuntos
Hemodinâmica , Hipóxia/fisiopatologia , Receptores Adrenérgicos beta 2/fisiologia , Animais , Pressão Arterial , Atenolol/farmacologia , Catecolaminas/sangue , Frequência Cardíaca , Masculino , Propanolaminas/farmacologia , Propranolol/farmacologia , Ratos , Ratos Sprague-Dawley , Resistência Vascular
8.
Cell Mol Gastroenterol Hepatol ; 10(4): 763-778, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32629118

RESUMO

BACKGROUND & AIMS: Epidemiological and animal studies have indicated an inverse correlation between the rising prevalence of obesity and metabolic syndrome and exposure to helminths. Whether helminth-induced immune response contributes to microbiota remodeling in obesity remains unknown. The aim of this study is to explore the immune-regulatory role of helminth in the prevention of HFD-induced obesity through remodeling gut microbiome. METHODS: C57BL/6J WT and STAT6-/- mice were infected with Heligmosomoides polygyrus and followed by high fat diet (HFD) feeding for 6 weeks. The host immune response, body weight, and fecal microbiota composition were analyzed. We used adoptive transfer of M2 macrophages and microbiota transplantation approaches to determine the impact of these factors on HFD-obesity. We also examined stool microbiota composition and short chain fatty acids (SCFAs) concentration and determined the expression of SCFA-relevant receptors in the recipient mice. RESULTS: Helminth infection of STAT6-/- (Th2-deficient) mice and adoptive transfer of helminth-induced alternatively activated (M2) macrophages demonstrated that the helminth-associated Th2 immune response plays an important role in the protection against obesity and induces changes in microbiota composition. Microbiota transplantation showed that helminth-induced, Th2-dependent alterations of the gut microbiota are sufficient to confer protection against obesity. Collectively, these results indicate that helminth infection protects against HFD-induced obesity by Th2-dependent, M2 macrophage-mediated alterations of the intestinal microbiota. CONCLUSION: Our findings provide new mechanistic insights into the complex interplay between helminth infection, the immune system and the gut microbiota in a HFD-induced obesity model and holds promise for gut microbiome-targeted immunotherapy in obesity prevention.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Microbioma Gastrointestinal , Nematospiroides dubius/imunologia , Obesidade/prevenção & controle , Animais , Células Cultivadas , Camundongos Endogâmicos C57BL , Obesidade/etiologia , Obesidade/imunologia , Fatores de Proteção , Infecções por Strongylida/imunologia
9.
PLoS One ; 13(9): e0203701, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30208088

RESUMO

This experiment was conducted to investigate the effect of exposure to high altitude and low oxygen on intestinal microbial communities using mice as an animal model. Fecal microbiota from mice housed in a control environment representing 2,200 meters (NC group) above sea level with 16% Oxygen and mice that were placed in a hypobaric chamber representing 5000 meters (HC group) above sea level with 11% Oxygen for 30 days, were analyzed by the HiSeq Illumina sequencing platform. The results showed a significant difference in beta diversity observed between the two groups, while no significant difference was observed in alpha diversity. Compared with the NC group, the relative abundance of class Epsilonproteobacteria, phlym Actinobacteria, class Erysipelotrichia and genus Helicobacter were significantly lower (P<0.05), while the relative abundance of genus Alistipes was increased in the HC group; Phenotypic analysis showed no significant difference in aerobic, anaerobic, facultatively anaerobic, potentially pathogenic, stress tolerant, mobile element, biofilms formation, gram negative and gram positive between HC group and NC group; Functional analysis results showed significant differences in 34 gene functional metabolic pathways (carbohydrate digestion and absorption, energy metabolism, arachidonic acid metabolism, flavonoid biosynthesis, RIG-I-like receptor signaling pathway, etc) between HC group and NC group. Together, these findings suggest that exposure to high altitude and low oxygen had the potential to change the intestinal microbial communities, which potentially may modulate metabolic processes in mice.


Assuntos
Microbioma Gastrointestinal/fisiologia , Hipóxia , Altitude , Animais , Bactérias/genética , Bactérias/isolamento & purificação , Fezes/microbiologia , Feminino , Hematócrito , Hemoglobinas/análise , Sequenciamento de Nucleotídeos em Larga Escala , Redes e Vias Metabólicas/genética , Camundongos , Camundongos Endogâmicos BALB C , Fenótipo , RNA Ribossômico 16S/química , RNA Ribossômico 16S/genética , RNA Ribossômico 16S/metabolismo , Análise de Sequência de DNA
10.
Sheng Li Xue Bao ; 70(2): 115-122, 2018 Apr 25.
Artigo em Chinês | MEDLINE | ID: mdl-29691575

RESUMO

The present study is aimed to explore the effects of endogenous carbon monoxide on the ischemia-reperfusion in rats. Wistar rats were intraperitoneally injected with protoporphyrin cobalt chloride (CoPP, an endogenous carbon monoxide agonist, 5 mg/kg), zinc protoporphyrin (ZnPP, an endogenous carbon monoxide inhibitor, 5 mg/kg) or saline. Twenty-four hours after injection, the myocardial ischemia-reperfusion model was made by Langendorff isolated cardiac perfusion system, and cardiac function parameters were collected. Myocardial cGMP content was measured by ELISA, and the endogenous carbon monoxide in plasma and myocardial enzymes in perfusate at 10 min after reperfusion were measured by colorimetry. The results showed that before ischemia the cardiac functions of CoPP, ZnPP and control groups were stable, and there were no significant differences. After reperfusion, cardiac functions had significant differences among the three groups (P < 0.05). Compared with pre-ischemia, the cardiac function decreased and obvious cardiac arrest was shown in control and ZnPP groups, while the cardiac function in CoPP group did not change significantly, maintaining a relatively stable level. At the same time, three groups' carbon monoxide level, myocardial enzymology and the cardiac function recovery time after reperfusion also had significant differences (P < 0.05). Compared with those in control group, recovery after reperfusion was faster, activities of creatine kinase and lactic dehydrogenase were significantly decreased, plasma CO and myocardial cGMP contents were significantly increased in CoPP group, while these changes were completely opposite in ZnPP group. It is concluded that endogenous carbon monoxide can maintain cardiac function, shorten the time of cardiac function recovery, and play a protective role in cardiac ischemia-reperfusion.


Assuntos
Monóxido de Carbono/sangue , Traumatismo por Reperfusão Miocárdica , Animais , Monóxido de Carbono/agonistas , Monóxido de Carbono/antagonistas & inibidores , Creatina Quinase/metabolismo , GMP Cíclico/metabolismo , Coração , L-Lactato Desidrogenase/metabolismo , Miocárdio/enzimologia , Protoporfirinas/administração & dosagem , Ratos , Ratos Sprague-Dawley , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...