Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Immun Inflamm Dis ; 12(4): e1237, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38577984

RESUMO

BACKGROUND: Severe myocarditis is often accompanied by cardiac fibrosis, but the underlying mechanism has not been fully elucidated. CXCL4 is a chemokine that has been reported to have pro-inflammatory and profibrotic functions. The exact role of CXCL4 in cardiac fibrosis remains unclear. METHODS: Viral myocarditis (VMC) models were induced by intraperitoneal injection of Coxsackie B Type 3 (CVB3). In vivo, CVB3 (100 TCID50) and CVB3-AMG487 (CVB3: 100 TCID50; AMG487: 5 mg/kg) combination were administered in the VMC and VMC+AMG487 groups, respectively. Hematoxylin and eosin staining, severity score, Masson staining, and immunofluorescence staining were performed to measure myocardial morphology in VMC. Enzyme-linked immunosorbent assay (ELISA) and quantitative reverse transcription polymerase chain reaction (qRT-PCR) were performed to quantify inflammatory factors (IL-1ß, IL-6, TNF-α, and CXCL4). Aspartate aminotransferase (AST), lactate dehydrogenase (LDH), and creatine kinase-myocardial band (CK-MB) levels were analyzed by commercial kits. CXCL4, CXCR3B, α-SMA, TGF-ß1, Collagen I, and Collagen III were determined by Western blot and immunofluorescence staining. RESULTS: In vivo, CVB3-AMG487 reduced cardiac injury, α-SMA, Collagen I and Collagen III levels, and collagen deposition in VMC+AMG487 group. Additionally, compared with VMC group, VMC+AMG group decreased the levels of inflammatory factors (IL-1ß, IL-6, and TNF-α). In vitro, CXCL4/CXCR3B axis activation TGF-ß1/Smad2/3 pathway promote mice cardiac fibroblasts differentiation. CONCLUSION: CXCL4 acts as a profibrotic factor in TGF-ß1/Smad2/3 pathway-induced cardiac fibroblast activation and ECM synthesis, and eventually progresses to cardiac fibrosis. Therefore, our findings revealed the role of CXCL4 in VMC and unveiled its underlying mechanism. CXCL4 appears to be a potential target for the treatment of VMC.


Assuntos
Acetamidas , Infecções por Coxsackievirus , Miocardite , Pirimidinonas , Camundongos , Animais , Fator de Crescimento Transformador beta1/metabolismo , Fator de Necrose Tumoral alfa , Interleucina-6 , Colágeno , Fibrose
2.
Inflammation ; 46(4): 1305-1317, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37036563

RESUMO

Soluble interleukin 1 receptor-like 1 (sST2) is a novel predictor of poor outcomes, which is involved in inflammatory response and fibrosis of myocarditis. Cellular senescence is a state of irreversible cell cycle arrest. Studies have shown that senescence of myofibroblasts can limit or reduce cardiac fibrosis. However, the molecular mechanism of sST2 regulating cellular senescence is still unclear. Here, we investigate the role of sST2 on cellular senescence in cardiac fibrosis. Our results found that sST2 was upregulated in coxsackievirus group B type 3 (CVB3)-induced viral myocarditis (VMC), which correlated with the expression of senescence markers. In vitro, sST2 activated TGFß signaling through the phosphorylation of the SMAD complex to induce mouse cardiac fibroblast (MCF) activation and inhibit cellular senescence by the Sirt1/p53/p21 signaling pathway. In vivo, anti-ST2 mAb attenuated CVB3-induced cardiac fibrosis. Our findings elucidate a crucial mechanism underlying through which sST2 inhibits cellular senescence and regulates MCF activation, providing a potential treatment strategy for cardiac fibrosis.


Assuntos
Miocardite , Camundongos , Animais , Miocardite/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Sirtuína 1/metabolismo , Fibrose
3.
FASEB J ; 36(9): e22472, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35959877

RESUMO

The mechanisms underlying acute kidney injury (AKI) and chronic kidney disease (CKD) progression include interstitial inflammation, cellular senescence, and oxidative stress (OS). Although vanin-1 (VNN1) plays an important role in OS, its contribution to the AKI-CKD transition remains unknown. Here, we explored the roles and mechanisms of VNN1 in the progression of the AKI-CKD transition. We observed that VNN1 expression was upregulated after ischemia/reperfusion (I/R) injury and high VNN1 expression levels were associated with poor renal repair after I/R injury. In VNN1 knockout (KO) mice, recovery of serum creatinine and blood urea nitrogen levels after I/R injury was accelerated and renal fibrosis was inhibited after severe I/R injury. Furthermore, in VNN1 KO mice, senescence of renal tubular cells was inhibited after severe I/R injury, as assessed by P16 expression and SA-ß-Gal assays. However, our results also revealed that VNN1 KO renal tubular cells did not resist senescence when OS was blocked. To elucidate the mechanism underlying VNN1-mediated regulation of senescence during the AKI-CKD transition, retinoblastoma 1 (RB1) was identified as a potential target. Our results suggest that the reduced senescence in VNN1 KO renal tubular cells was caused by suppressed RB1 expression and phosphorylation. Collectively, our results unveil a novel molecular mechanism by which VNN1 promotes AKI-CKD transition via inducing senescence of renal tubular cells by activating RB1 expression and phosphorylation after severe renal injury. The present study proposes a new strategy for designing therapies wherein VNN1 can be targeted to obstruct the AKI-CKD transition.


Assuntos
Injúria Renal Aguda , Insuficiência Renal Crônica , Traumatismo por Reperfusão , Injúria Renal Aguda/genética , Injúria Renal Aguda/metabolismo , Animais , Senescência Celular , Fibrose , Rim/metabolismo , Camundongos , Camundongos Knockout , Insuficiência Renal Crônica/metabolismo , Traumatismo por Reperfusão/metabolismo
4.
World J Emerg Surg ; 15(1): 33, 2020 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-32414390

RESUMO

BACKGROUND: A novel coronavirus pneumonia outbreak began in Wuhan, Hubei Province, in December 2019; the outbreak was caused by a novel coronavirus previously never observed in humans. China has imposed the strictest quarantine and closed management measures in history to control the spread of the disease. However, a high level of evidence to support the surgical management of potential trauma patients during the novel coronavirus outbreak is still lacking. To regulate the emergency treatment of trauma patients during the outbreak, we drafted this paper from a trauma surgeon perspective according to practical experience in Wuhan. MAIN BODY: The article illustrates the general principles for the triage and evaluation of trauma patients during the outbreak of COVID-19, indications for emergency surgery, and infection prevention and control for medical personnel, providing a practical algorithm for trauma care providers during the outbreak period. CONCLUSIONS: The measures of emergency trauma care that we have provided can protect the medical personnel involved in emergency care and ensure the timeliness of effective interventions during the outbreak of COVID-19.


Assuntos
Infecções por Coronavirus , Transmissão de Doença Infecciosa/prevenção & controle , Controle de Infecções/normas , Pandemias , Pneumonia Viral , Ferimentos e Lesões/diagnóstico , Ferimentos e Lesões/terapia , Algoritmos , Anestesia/normas , COVID-19 , China , Infecções por Coronavirus/epidemiologia , Infecções por Coronavirus/prevenção & controle , Infecções por Coronavirus/transmissão , Emergências , Unidades Hospitalares/normas , Humanos , Pandemias/prevenção & controle , Assistência Perioperatória/normas , Equipamento de Proteção Individual/normas , Pneumonia Viral/epidemiologia , Pneumonia Viral/prevenção & controle , Pneumonia Viral/transmissão , Procedimentos Cirúrgicos Operatórios/normas , Tomografia Computadorizada por Raios X/normas , Triagem/normas
5.
Eur J Immunol ; 50(6): 795-808, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32068249

RESUMO

Resident cardiac macrophages play important roles in homeostasis, maintenance of cardiac function, and tissue repair. After cardiac injury, monocytes infiltrate the tissue, undergo phenotypic and functional changes, and are involved in inflammatory injury and functional remodelling. However, the fate of cardiac infiltrating/polarized macrophages and the relationship between these cells and resident cardiac macrophage replenishment following injury remain unclear. Our results showed that angiotensin II induces cardiac fibroblast transdifferentiation into cardiac myofibroblasts (MFBs). In cocultures with MFBs and murine macrophages, the MFBs promoted macrophage polarization to M1 phenotype, followed by selective apoptosis, which was associated with TNF/TNFR1 axis and independent of NO production. Surprisingly, after 36 h of coculture, the surviving macrophages were converted to M2 phenotype and settled in heart, which was dependent on leptin produced by MFBs or polarized macrophages via the PI3K or Akt pathway. CCR2+ CD45.2+ cells adoptively transferred into CD45.1+ mice with viral myocarditis, differentiated into CD45.2+ CCR2+ CX3CR1+ M2 cells during the resolution of inflammation and settled within the heart. Our data highlight a novel mechanism related to the renewal or replenishment of cardiac resident macrophages following cardiac injury; and suggest that transdifferentiation of cardiac fibroblasts may promote the resolution of inflammation.


Assuntos
Transdiferenciação Celular/imunologia , Fibroblastos/imunologia , Macrófagos/imunologia , Miocardite/imunologia , Miocárdio/imunologia , Transdução de Sinais/imunologia , Animais , Transdiferenciação Celular/genética , Fibroblastos/patologia , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Miocardite/genética , Miocardite/patologia , Miocárdio/patologia , Transdução de Sinais/genética
6.
Cell Immunol ; 347: 104025, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31837749

RESUMO

B10 cells, a specific subset of regulatory B cells, are capable of regulating immune response and restricting inflammation and autoimmune disease progression by producing IL-10. B10 cells frequently change significantly during inflammation and autoimmunity. However, how B10 cell populations change in viral myocarditis (VMC) remains unclear. Therefore, this work was conducted to clarify the changes in B10 cells and their potential mechanisms. Our results showed that the B10 cell frequency significantly changed in the VMC model. Changes in prostaglandin E2 (PGE2) levels in VMC model hearts were consistent with B10 expansion. PGE2 induced B10 cell expansion via the MAPKs/AKT-AP1 axis or AhR signaling. Additionally, PGE2-pretreated B10 cells inhibited naïve CD4+ T cell differentiation into Th17 cells. In vivo, PGE2 treatment or adoptive B10 cell transfer significantly restricted VMC development. Our results provide sufficient evidence that PGE2-induced B10 cell expansion may become a promising therapeutic approach for VMC and acute inflammatory injury.


Assuntos
Linfócitos B Reguladores/imunologia , Dinoprostona/farmacologia , Infecções por Enterovirus/patologia , Interleucina-10/imunologia , Miocardite/prevenção & controle , Células Th17/citologia , Transferência Adotiva , Animais , Subpopulações de Linfócitos B/imunologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Dinoprostona/sangue , Enterovirus Humano B , Ativação Linfocitária/imunologia , Contagem de Linfócitos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Miocardite/tratamento farmacológico , Miocardite/virologia , Miocárdio/imunologia , Peptídeos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/imunologia , Células Th17/imunologia
7.
J Clin Immunol ; 40(1): 147-157, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31749032

RESUMO

miR-608 has been indicated to play an important role in the pathogenesis of various inflammation-related diseases, including sepsis and several types of cancers. However, there is little information about the underlying mechanism, especially in inflammatory cells. In this study, an hsa-miR-608-inhibition cell model was constructed in U937 cells using a lentivirus, and gene expression profiles were determined by a cDNA microarray. Altogether, 682 genes showed a difference greater than 1.2-fold, including 184 genes downregulated and 498 genes upregulated. Among these genes, one potential miR-608-target gene, ELANE, was further investigated. A positive relationship between the expression of miR-608 and that of ELANE was found both in vivo and in vitro. In addition, decreased expression of miR-608 resulted in overexpression of ELANE at both the mRNA and protein levels. Cotransfection of HEK293T cells with a miR-608 mimic inhibited reporter activity, and mutation of the miRNA seed sequences abolished the repression of reporter activity. These results suggest that miR-608 is an important posttranscriptional regulator of ELANE expression in human monocytes and may play an important role in the process of inflammation. miR-608 and neutrophil elastase may be novel targets for the diagnosis or treatment of sepsis.


Assuntos
Anti-Inflamatórios/metabolismo , Inflamação/metabolismo , Elastase de Leucócito/metabolismo , MicroRNAs/metabolismo , Monócitos/metabolismo , Adulto , Linhagem Celular , Linhagem Celular Tumoral , Regulação para Baixo/fisiologia , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica/fisiologia , Células HEK293 , Humanos , RNA Mensageiro/metabolismo , Transdução de Sinais/fisiologia , Células THP-1 , Células U937 , Regulação para Cima/fisiologia , Adulto Jovem
8.
World J Emerg Surg ; 14: 11, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30918528

RESUMO

Background: Patients suffering from major trauma often experience complications such as sepsis. The early recognition of patients at high risk of sepsis after trauma is critical for precision therapy. We aimed to derive and validate a novel predictive score for sepsis risk using electronic medical record (EMR) data following trauma. Materials and methods: Clinical and laboratory variables of 684 trauma patients within 24 h after admission were collected, including 411 patients in the training cohort and 273 in the validation cohort. The least absolute shrinkage and selection operator (LASSO) technique was adopted to identify variables contributing to the early prediction of traumatic sepsis. Then, we constructed a traumatic sepsis score (TSS) using a logistic regression model based on the variables selected in the LASSO analysis. Moreover, we evaluated the discrimination and calibration of the TSS using the area under the curve (AUC) and the Hosmer-Lemeshow (H-L) goodness-of-fit test. Results: Based on the LASSO, seven variables (injury severity score, Glasgow Coma Scale, temperature, heart rate, albumin, international normalized ratio, and C-reaction protein) were selected for construction of the TSS. Our results indicated that the incidence of sepsis after trauma increased with an increasing TSS (Ptrend = 7.44 × 10-21 for the training cohort and Ptrend = 1.16 × 10-13 for the validation cohort). The areas under the receiver operating characteristic (ROC) curve of TSS were 0.799 (0.757-0.837) and 0.790 (0.736-0.836) for the training and validation datasets, respectively. The discriminatory power of our model was superior to that of a single variable and the sequential organ failure assessment (SOFA) score (P < 0.001). Moreover, the TSS was well calibrated (P > 0.05). Conclusions: We developed and validated a novel TSS with good discriminatory power and calibration for the prediction of sepsis risk in trauma patients based on the EMR data.


Assuntos
Valor Preditivo dos Testes , Sepse/diagnóstico , Índice de Gravidade de Doença , Adolescente , Adulto , Área Sob a Curva , Feminino , Humanos , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Escores de Disfunção Orgânica , Prognóstico , Estudos Prospectivos , Curva ROC , Medição de Risco/métodos , Medição de Risco/normas , Estatísticas não Paramétricas , Ferimentos e Lesões/diagnóstico , Ferimentos e Lesões/fisiopatologia
9.
J Trauma Acute Care Surg ; 86(3): 440-447, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30489503

RESUMO

BACKGROUND: Genetic backgrounds have been recognized as significant determinants of susceptibility to sepsis. CXC chemokines play a significant role in innate immunity against infectious diseases. Genetic polymorphisms of CXC chemokine genes have been widely studied in inflammatory and infectious diseases but not in sepsis. Thus, we aimed to investigate the clinical relevance of CXC chemokine gene polymorphisms and susceptibility to sepsis in a traumatically injured population. METHODS: Thirteen tag single nucleotide polymorphisms were selected from CXC chemokine genes using a multimarker tagging algorithm in the Tagger software. Three independent cohorts of injured patients (n = 1700) were prospectively recruited. Selected single nucleotide polymorphisms were genotyped using an improved multiplex ligation detection reaction method. Cytokine production in lipopolysaccharide-stimulated whole blood was measured using an enzyme-linked immunosorbent assay. RESULTS: Among the 13 tag single nucleotide polymorphisms, four single nucleotide polymorphisms (rs1429638, rs266087, rs2297630, and rs2839693) were significantly associated with the susceptibility to sepsis, and three (rs3117604, rs1429638, and rs4074) were significantly associated with an increased multiple organ dysfunction score in the derivation cohort. However, only the clinical relevance of rs1429638 and rs266087 was confirmed in the validation cohorts. In addition, rs2297630 was significantly associated with interleukin 6 production. CONCLUSION: The rs1429638 polymorphism in the CXCL1 gene and the rs2297630 polymorphism in the CXCL12 gene were associated with altered susceptibility to sepsis and might be used as important genetic markers to assess the risks of sepsis in trauma patients. LEVEL OF EVIDENCE: Prognostic and epidemiologic study, level II.


Assuntos
Quimiocina CXCL12/genética , Quimiocina CXCL1/genética , Polimorfismo de Nucleotídeo Único , Sepse/genética , Ferimentos e Lesões , Adulto , China , Feminino , Predisposição Genética para Doença , Genótipo , Humanos , Masculino , Estudos Prospectivos , Fatores de Risco
10.
World J Emerg Surg ; 13: 52, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30479651

RESUMO

Background: Previous study revealed that rs2232618 polymorphism (Phe436Leu) within LBP gene is a functional variant and associated with susceptibility of sepsis in traumatic patients. Our aim was to confirm the reported association by enlarging the population sample size and perform a meta-analysis to find additional evidence. Methods: Traumatic patients from Southwest (n = 1296) and Southeast (n = 445) of China were enrolled in our study. After genotyping, the relationship between rs2232618 and the risk of sepsis was analyzed. Furthermore, we proceeded with a comprehensive literature search and meta-analysis to determine whether the rs2232618 polymorphism conferred susceptibility to sepsis. Results: Significance correlation was observed between rs2232618 and risk of sepsis in Southwest patients (P = 0.002 for the dominant model, P = 0.006 for the recessive model). The association was confirmed in Southeast cohort (P = 0.005 for the dominant model) and overall combined cohorts (P = 4.5 × 10-4, P = 0.041 for the dominant and recessive model). Multiple logistical regression analyses suggested that rs2232618 polymorphism was related to higher risk of sepsis (OR = 1.77, 95% CI = 1.26-2.48, P = 0.001 in Southwest patients; OR = 2.11, 95% CI = 1.24-3.58, P = 0.006 in Southeast cohort; OR = 1.54, 95% CI = 1.34-2.08, P = 0.006 in overall cohort). Furthermore, meta-analysis of four studies (including the present study) confirmed that rs2232618 within LBP increased the risk of sepsis (OR = 1.75, P < 0.001 for the dominant model; OR = 6.08, P = 0.003 for the recessive model; OR = 2.72, P < 0.001 for the allelic model). Conclusions: The results from our replication study and meta-analysis provided firm evidence that rs2232618T allele significantly increased the risk of sepsis.


Assuntos
Predisposição Genética para Doença/genética , Polimorfismo Genético/genética , Sepse/etiologia , Sepse/genética , Ferimentos e Lesões/complicações , Proteínas de Fase Aguda , Proteínas de Transporte/sangue , China , Frequência do Gene , Genótipo , Humanos , Glicoproteínas de Membrana/sangue , Fatores de Risco , Sepse/sangue , Sepse/fisiopatologia , Ferimentos e Lesões/sangue , Ferimentos e Lesões/fisiopatologia
11.
Cell Biol Int ; 42(9): 1160-1169, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29741224

RESUMO

The high-mobility group box-1 (HMGB1), as a highly conserved ubiquitous DNA-binding protein, has been widely studied in various diseases, including inflammation and tumor; however, fewer studies were focused on the mechanisms controlling HMGB1 release compared with the function of HMGB1. Previous studies have proven that ANG II can act as a pro-inflammatory cytokine, both of HMGB1 and ANG II were significantly upregulated in autoimmune diseases; however, the exact role of ANG II in regulating HMGB1 release have not been shown. The present study was to define the effects of ANG II on macrophages and the possible mechanisms in controlling HMGB1 release. Our results showed that ANG II can induce M1 macrophage polarization through upregulated the expression of HMGB1 and caused acetylation of HMGB1 and release via its dissociation from SIRT1, which in a positive feedback upregulates ANG II. Subsequently, HMGB1 inhibitors can reduce the ANG II-elicited polarize of macrophage. Meanwhile, we show that JAK/STAT pathways play an essential role in ANG II-induced HMGB1 nuclear translocation, JAK/STAT specific inhibitors can inhibit ANG II-induced HMGB1 expression. Taken together, our results provide a novel evidence that HMGB1 play a critical role in ANG II mediated macrophage polarization, and we suggest that ANG II mediated HMGB1 release via dissociation from SIRT1, induce hyperacetylation of HMGB1, thus for subsequent release, suggesting that the angiotensin II receptor antagonist is a potential drug target for inhibiting HMGB1 release in inflammation diseases.


Assuntos
Angiotensina II/metabolismo , Proteína HMGB1/metabolismo , Acetilação , Animais , Polaridade Celular/fisiologia , Citocinas/sangue , Citocinas/metabolismo , Proteína HMGB1/biossíntese , Proteína HMGB1/genética , Inflamação/metabolismo , Macrófagos Peritoneais/metabolismo , Camundongos , Transporte Proteico , Células RAW 264.7 , Transdução de Sinais , Sirtuína 1/metabolismo
12.
Int Immunopharmacol ; 56: 277-284, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29414662

RESUMO

High-mobility group box 1 (HMGB1), an important inflammatory factor, plays significant roles in CD4+T cell differentiation, cancer and autoimmune disease development. Our previous data have demonstrated that HMGB1 contributes to macrophage reprogramming and is involved in experimental autoimmune myocarditis (EAM) development. In contrast to the well-explored function of HMGB1, little is known about the nuclear function. Whether HMGB1 can serve as an architectural factor and control functional skewing of macrophages remains unclear. Therefore, the present work was performed to address the above speculation. The adenovirus-mediated shRNA (Ad-shRNA) was employed to knock down HMGB1 in RAW264.7 and monocytes/macrophages of EAM mice. Our data showed that in vitro HMGB1 silencing limited functional skewing of macrophages and down-regulated inflammatory factors secretion, which can't be reversed by the exogenous HMGB1. In M1 polarization system, the phosphorylations of NF-κB, p38 and Erk1/2 were inhibited following HMGB1 silencing. In vivo, HMGB1 silencing could effectively ameliorate EAM development. Our data suggest that HMGB1 may be a checkpoint nuclear factor of macrophage reprogramming. Our findings also provide an exciting therapeutic method for inflammatory disorders.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Proteína HMGB1/metabolismo , Macrófagos/imunologia , Esclerose Múltipla/imunologia , Animais , Autoantígenos/imunologia , Miosinas Cardíacas/imunologia , Diferenciação Celular , Reprogramação Celular , Citocinas/metabolismo , Modelos Animais de Doenças , Progressão da Doença , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteína HMGB1/genética , Humanos , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Endogâmicos BALB C , Cadeias Pesadas de Miosina/imunologia , NF-kappa B/metabolismo , Células RAW 264.7 , RNA Interferente Pequeno/genética , Células Th1/imunologia
13.
J Leukoc Biol ; 103(4): 719-730, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29350825

RESUMO

Macrophage, a highly plastic population, is widely distributed. Macrophage functions are settled and acquired polarization programs in response to microenvironmental signals and involved in many inflammatory disorders, such as experimental autoimmune myocarditis (EAM). Phenotypic and functional changes in macrophage are considered as an important determinant of disease progression and/or regression. Angiotensin II (ANG II), as a powerful proinflammatory factor, plays critical roles in inflammatory diseases and macrophage recruitment. It remains unclear whether ANG II contributed to the functional skewing of cardiac infiltrated monocytes/macrophage and involved in EAM development. Therefore, the present work was to address the above questions. Our data showed that ANG II contributed to CD11b+ Ly6Chi (CD11b+ Ly6G- Ly6C+ ) cells reprogramming into M1-like macrophage through Erk1/2 or p38/Stat3 pathway and the reprogramming M1-like cells promoted Th17 cells expansion; abrogation of ANG II-AT1 R axis significantly ameliorated cardiac injury. The present work first demonstrated a novel immune regulation role of ANG II; ANG II, as a powerful immune factor, promoted CD11b+ Ly6Chi inflammatory cells reprogramming into M1-like macrophage and involved in inflammatory disorders development; our results also indicated that ANG II may be a potential therapeutic target for inflammatory diseases.


Assuntos
Angiotensina II/farmacologia , Doenças Autoimunes/tratamento farmacológico , Macrófagos/imunologia , Monócitos/imunologia , Miocardite/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Animais , Antígenos Ly/metabolismo , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Doenças Autoimunes/patologia , Antígenos CD11/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Miocardite/imunologia , Miocardite/metabolismo , Miocardite/patologia , Fator de Transcrição STAT3/metabolismo , Vasoconstritores/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
14.
Microbiol Immunol ; 61(12): 539-546, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29052263

RESUMO

Angiotensin II (ANG II) plays critical roles in modulation of circulatory homeostasis and activation of innate and adaptive immunity and has also been implicated in several mouse models of autoimmune disease. However, how ANG II regulates macrophages and is involved in development of experimental autoimmune myocarditis (EAM) remains unclear. Therefore, the present study aimed to address the above question and explore possible mechanisms. EAM was induced in BALB/c mice. ANG II was quantitated by ELISA and hematoxylin and eosin staining was employed to analyze pathological changes and macrophage infiltration. The chemotactic ability of ANG II was assessed by using a Transwell system. It was found that ANG II is up-regulated in serum and heart tissues of mice with EAM and that ANG II significantly drives monocyte/macrophage infiltration through the C-C chemokine receptor 2/5 (CCR2/5) axis. CCR2/5 antagonists and ANG II receptor inhibitor could all abrogate monocyte/macrophage infiltration and ameliorate development of EAM. Our results have firstly identified a novel function of ANG II: that it is a critical chemokine for monocyte/macrophage recruitment. Furthermore, our results indicate that ANG II is a potential candidate for treatment of inflammatory diseases.


Assuntos
Angiotensina II/imunologia , Doenças Autoimunes/imunologia , Macrófagos/imunologia , Monócitos/imunologia , Miocardite/imunologia , Receptores CCR2/imunologia , Receptores CCR5/imunologia , Angiotensina II/genética , Animais , Doenças Autoimunes/genética , Progressão da Doença , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Miocardite/genética , Receptores CCR2/genética , Receptores CCR5/genética
15.
Biotechnol Biofuels ; 10: 181, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28702086

RESUMO

BACKGROUND: The biomass yield of Chlorella PY-ZU1 drastically increased when cultivated under high CO2 condition compared with that cultivated under air condition. However, less attention has been given to the microalgae photosynthetic mechanisms response to different CO2 concentrations. The genetic reasons for the higher growth rate, CO2 fixation rate, and photosynthetic efficiency of microalgal cells under higher CO2 concentration have not been clearly defined yet. RESULTS: In this study, the Illumina sequencing and de novo transcriptome assembly of Chlorella PY-ZU1 cells cultivated under 15% CO2 were performed and compared with those of cells grown under air. It was found that carbonic anhydrase (CAs, enzyme for interconversion of bicarbonate to CO2) dramatically decreased to near 0 in 15% CO2-grown cells, which indicated that CO2 molecules directly permeated into cells under high CO2 stress without CO2-concentrating mechanism. Extrapolating from the growth conditions and quantitative Real-Time PCR of CCM-related genes, the Km (CO2) (the minimum intracellular CO2 concentration that rubisco required) of Chlorella PY-ZU1 might be in the range of 80-192 µM. More adenosine triphosphates was saved for carbon fixation-related pathways. The transcript abundance of rubisco (the most important enzyme of CO2 fixation reaction) was 16.3 times higher in 15% CO2-grown cells than that under air. Besides, the transcript abundances of most key genes involved in carbon fixation pathways were also enhanced in 15% CO2-grown cells. CONCLUSIONS: Carbon fixation and nitrogen metabolism are the two most important metabolisms in the photosynthetic cells. These genes related to the two most metabolisms with significantly differential expressions were beneficial for microalgal growth (2.85 g L-1) under 15% CO2 concentration. Considering the micro and macro growth phenomena of Chlorella PY-ZU1 under different concentrations of CO2 (0.04-60%), CO2 transport pathways responses to different CO2 (0.04-60%) concentrations was reconstructed.

16.
Bioresour Technol ; 238: 650-656, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28486198

RESUMO

Spirulina sp. was mutated by γ-rays from 60Co nuclear irradiation to improve growth and CO2 fixation rate under 15vol.% CO2 (in flue gas from a power plant). Mutants with enhanced growth phenotype were obtained, with the best strain exhibiting 310% increment in biomass yield on day 4. The mutant was then domesticated with elevated CO2 concentration, and the biomass yield increased by 500% after domestication under 15vol.% CO2, with stable inheritance. Ultrastructure of Spirulina sp. shows that the fractal dimension of Spirulina cells decreased by 23% after mutation. Pore size in the cell wall of Spirulina mutant increased by 33% after 15vol.% CO2 domestication. This characteristic facilitated the direct penetration of CO2 into cells, thus improving CO2 biofixation rate.


Assuntos
Spirulina/genética , Biomassa , Reatores Biológicos , Dióxido de Carbono , Mutação , Centrais Elétricas
17.
Mil Med Res ; 3: 37, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27980800

RESUMO

BACKGROUND: People rapidly ascending to high altitudes (>2500 m) may suffer from acute mountain sickness (AMS). The association between smoking and AMS risk remains unclear. Therefore, we performed a meta-analysis to evaluate the association between smoking and AMS risk. METHODS: The association between smoking and AMS risk was determined according to predefined criteria established by our team. Meta-analysis was conducted according to the PRISMA guidelines. We included all relevant studies listed in the PubMed and Embase databases as of September 2015 in this meta-analysis and performed systemic searches using the terms "smoking", "acute mountain sickness" and "risk factor". The included studies were required to provide clear explanations regarding their definitions of smoking, the final altitudes reached by their participants and the diagnostic criteria used to diagnose AMS. Odds ratios (ORs) were used to evaluate the association between smoking and AMS risk across the studies, and the Q statistic was used to test OR heterogeneity, which was considered significant when P < 0.05. We also computed 95% confidence intervals (CIs). Data extracted from the articles were analyzed with Review Manager 5.3 (Cochrane Collaboration, Oxford, UK). RESULTS: We used seven case-control studies including 694 smoking patients and 1986 non-smoking controls to analyze the association between smoking and AMS risk. We observed a significant association between AMS and smoking (OR = 0.71, 95% CI 0.52-0.96, P = 0.03). CONCLUSIONS: We determined that smoking may protect against AMS development. However, we do not advise smoking to prevent AMS. More studies are necessary to confirm the role of smoking in AMS risk.


Assuntos
Doença da Altitude/complicações , Doença da Altitude/epidemiologia , Fumar/epidemiologia , Adulto , Feminino , Humanos , Masculino , Estudos Observacionais como Assunto , Fatores de Risco , Adulto Jovem
18.
Sci Rep ; 6: 21884, 2016 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-26899795

RESUMO

Macrophages can be reprogramming, such as the classical activated macrophage, M1 or alternative activated macrophages, M2 phenotype following the milieu danger signals, especially inflammatory factors. Macrophage reprogramming is now considered as a key determinant of disease development and/or regression. Experimental autoimmune myocarditis (EAM) is characterized by monocytes/macrophage infiltration, Th17 cells activation and inflammatory factors producing such as high mobility group box 1 (HMGB1). Whether infiltrated macrophages could be reprogramming in EAM? HMGB1 was associated with macrophage reprogramming? Our results clearly demonstrated that infiltrated macrophage was reprogrammed towards a proinflammatory M1-like phenotype and cardiac protection by monocytes/macrophages depletion or HMGB1 blockade in EAM; in vitro, HMGB1 facilitated macrophage reprogramming towards M1-like phenotype dependent on TLR4-PI3Kγ-Erk1/2 pathway; furthermore, the reprogramming M1-like macrophage promoted Th17 expansion. Therefore, we speculated that HMGB1 contributed EAM development via facilitating macrophage reprogramming towards M1-like phenotype except for directly modulating Th17 cells expansion.


Assuntos
Doenças Autoimunes/metabolismo , Proteína HMGB1/fisiologia , Macrófagos/fisiologia , Miocardite/metabolismo , Animais , Doenças Autoimunes/imunologia , Movimento Celular , Polaridade Celular , Proliferação de Células , Células Cultivadas , Sistema de Sinalização das MAP Quinases , Ativação de Macrófagos , Camundongos Endogâmicos BALB C , Miocardite/imunologia , Miocárdio/imunologia , Miocárdio/patologia , Fenótipo , Fatores de Proteção , Células Th17/imunologia
19.
Atherosclerosis ; 243(2): 421-8, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26520896

RESUMO

BACKGROUND: IFN-γ-producing Th17 cells have been implicated in autoimmune disorders, but their properties in humans are known only partially. The molecular mechanisms and external factors that govern IFN-γ-producing Th17-cell bias are incompletely understood. The present work was to clarify whether (i) IFN-γ-producing Th17 cells are present in the peripheral circulation of patients with coronary atherosclerosis (CA); (ii) high mobility group box (HMGB)1 in circulation is associated with IFN-γ-producing Th17-cell bias. METHODS: Thirty-six patients (17 females and 19 males; 45-84 years) diagnosed as having atherosclerosis after coronary angiography for suspected or known CA were included the study cohort. Samples of peripheral blood were collected from healthy volunteers and patients, and classical tests (flow cytometry, RT-qPCR) were used to measure blood components. RESULTS AND CONCLUSION: Our results clearly demonstrated that HMGB1 were up-regulated in different progressive CA patients: 5.38 ± 1.48 ng/ml, 6.30 ± 1.53 ng/ml and 5.86 ± 1.12 ng/ml vs1.45 ± 0.65 ng/ml for only atherosclerotic plaque (AP), atherosclerotic plaque and some plaque rupture, no thrombosis (PR), plaque rupture and accompanying thrombosis (TH) and volunteers, respectively, p < 0.05. The frequency of IFN-γ-producing Th17 cells was 2.33 ± 0.58%, 1.93 ± 0.2% and 2.21 ± 0.65% vs 0.38 ± 0.21% for AP, PR, TH and volunteers, p < 0.05, respectively. Furthermore, HMGB1 contributed to IFN-γ-producing Th17-cell bias by controlling expression of T-bet and RUNX3. We demonstrated, for the first time, that HMGB1 is a potential inducer of IFN-γ-producing Th17-cell bias, and that IFN-γ-producing Th17 cells might be one of the pathogenic factors in atherosclerosis.


Assuntos
Subunidade alfa 3 de Fator de Ligação ao Core/metabolismo , Doença da Artéria Coronariana/metabolismo , Trombose Coronária/metabolismo , Proteína HMGB1/metabolismo , Interferon gama/metabolismo , Proteínas com Domínio T/metabolismo , Células Th17/metabolismo , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Células Cultivadas , Subunidade alfa 3 de Fator de Ligação ao Core/genética , Doença da Artéria Coronariana/sangue , Doença da Artéria Coronariana/diagnóstico , Doença da Artéria Coronariana/genética , Doença da Artéria Coronariana/imunologia , Trombose Coronária/sangue , Trombose Coronária/diagnóstico , Trombose Coronária/genética , Trombose Coronária/imunologia , Progressão da Doença , Feminino , Proteína HMGB1/sangue , Proteína HMGB1/genética , Humanos , Interferon gama/sangue , Interferon gama/genética , Masculino , Pessoa de Meia-Idade , Placa Aterosclerótica , Interferência de RNA , Ruptura Espontânea , Índice de Gravidade de Doença , Transdução de Sinais , Proteínas com Domínio T/genética , Células Th17/imunologia , Transfecção
20.
Int J Clin Exp Pathol ; 8(12): 15940-6, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26884867

RESUMO

High mobility group box 1 (HMGB1), a non-histone nuclear protein, was associated with a variety of biological important processes, such as transcription, differentiation, extracellular signaling. As a cytokine or inflammatory mediator, more and more data showed that HMGB1 was involved in inflammatory diseases, cancers or autoimmune disease. However, few data focused on nucleic or cytoplasmic function of HMGB1. Therefore, the present study focused on cancer cells biological characteristics following HMGB1 silence. HMGB1 siRNAs were designed and chemically synthesized, and then transfected into the breast cancer cell line MCF-7 with lipofectamine 2000. The transcription and translation level of HMGB1 expression, proliferation, apoptosis, migration of MCF-7 were determined. The results demonstrated that HMGB1 silence inhibit invasion and migration and promote apoptosis of human breast cells; which indicated that HMGB1 silence might be a potential therapy targets.


Assuntos
Apoptose , Neoplasias da Mama/genética , Movimento Celular , Proteína HMGB1/genética , Interferência de RNA , RNA Interferente Pequeno/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Proteína HMGB1/metabolismo , Humanos , Células MCF-7 , Invasividade Neoplásica , Metástase Neoplásica , RNA Interferente Pequeno/metabolismo , Fatores de Tempo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA