Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 277
Filtrar
1.
J Clin Lab Anal ; 36(5): e24309, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35325494

RESUMO

BACKGROUND: Long noncoding RNA GAS5 (lnc-GAS5) is able to regulate macrophage M1 polarization and Th17 cell differentiation, also engaged in sepsis-induced inflammation and organ injury. This study aimed to further evaluate its linkage with Th1 cells and Th17 cells, as well as its clinical value in sepsis management. METHODS: About 101 sepsis patients were enrolled followed by peripheral blood mononuclear cell (PBMC) and serum samples collection. PBMC lnc-GAS5 was detected by RT-qPCR; Th1 cells and Th17 cells in PBMC CD4+ T cells were detected by flow cytometry; serum IFN-γ and IL-17A were detected by ELISA. Besides, PBMC lnc-GAS5 was also detected in 50 health controls (HCs). RESULTS: Lnc-GAS5 was reduced in sepsis patients than in HCs (p < 0.001), which also well-distinguished sepsis patients from HCs with AUC 0.860. Lnc-GAS5 did not relate to Th1 cells (p = 0.059) or IFN-γ (p = 0.192); while negatively linked with Th17 cells (p = 0.002) and IL-17A (p = 0.019) in sepsis patients. Interestingly, lnc-GAS5 negatively correlated with SOFA score (p = 0.001), SOFA-Respiratory system score (p = 0.001), SOFA-Coagulation score (p = 0.015), and SOFA-Renal system score (p = 0.026), but not SOFA-Liver score (p = 0.080), SOFA-Cardiovascular system score (p = 0.207) or SOFA-Nervous system score (p = 0.182) in sepsis patients. Furthermore, lnc-GAS5 was negatively related to CRP (p = 0.002) and APACHE II score (p = 0.004) in sepsis patients. Finally, lnc-GAS5 was decreased in dead sepsis patients compared to survivors (p = 0.007), which also distinguished sepsis deaths from survivors with AUC 0.713. CONCLUSION: Lnc-GAS5 relates to Th17 cells and serves as a potential biomarker for sepsis severity and mortality risk.


Assuntos
RNA Longo não Codificante , Sepse , Células Th17 , Biomarcadores , Humanos , Inflamação/imunologia , Interleucina-17/imunologia , Leucócitos Mononucleares/imunologia , Insuficiência de Múltiplos Órgãos/imunologia , Prognóstico , RNA Longo não Codificante/imunologia , Sepse/imunologia , Células Th17/imunologia
2.
Biomed Pharmacother ; 145: 112419, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34781146

RESUMO

Interleukin-6 (IL-6) is a multi-tasking cytokine that represents high activity in patients with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and cancer. High concentration of this pleiotropic cytokine accounts for hyperinflammation and cytokine storm, and is related to multi-organ failure in patients with SARS-CoV-2 induced disease. IL-6 promotes lymphopenia and increases C-reactive protein (CRP) in such cases. However, blockade of IL-6 is not a full-proof of complete response. Hypoxia, hypoxemia, aberrant angiogenesis and chronic inflammation are inter-related events occurring as a response to the SARS-CoV-2 stimulatory effect on high IL-6 activity. Taking both pro- and anti-inflammatory activities will make complex targeting IL-6 in patient with SARS-CoV-2 induced disease. The aim of this review was to discuss about interactions occurring within the body of patients with SARS-CoV-2 induced disease who are representing high IL-6 levels, and to determine whether IL-6 inhibition therapy is effective for such patients or not. We also address the interactions and targeted therapies in cancer patients who also have SARS-CoV-2 induced disease.


Assuntos
Tratamento Farmacológico da COVID-19 , COVID-19 , Inibidores de Checkpoint Imunológico/farmacologia , Interleucina-6 , Insuficiência de Múltiplos Órgãos , Neoplasias , Anticorpos Monoclonais Humanizados/farmacologia , COVID-19/complicações , COVID-19/imunologia , Humanos , Interleucina-6/antagonistas & inibidores , Interleucina-6/imunologia , Insuficiência de Múltiplos Órgãos/etiologia , Insuficiência de Múltiplos Órgãos/imunologia , Neoplasias/complicações , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , SARS-CoV-2
3.
Front Immunol ; 12: 742990, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34970255

RESUMO

Background: Human parvovirus B19 (B19) can cause acute hepatitis and is attributed to the high mortality of alcoholic hepatitis (AH). B19 infection is generally self-healing in previously healthy people, but it can cause fatal effects in some high-risk groups and increase its virulence and infectivity. Disseminated B19 infection-induced multiple organ dysfunction syndrome (MODS) in patients with AH has not been reported yet. Here, we described B19 viremia in an adult patient with AH accompanied by hemolytic anemia (HA), leading to disseminated infection and secondary MODS, as well as self-limiting B19 infections in seven nurses caring for him. Meanwhile, we reviewed the literature on AH and B19 infection. Case Presentation: A 43-year-old male patient with AH accompanied by HA was transferred to the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China, on March 31, 2021. After supportive treatment, his transaminase and bilirubin levels were reduced, but his anemia worsened. He received a red blood cell (RBC) infusion on April 9 for hemoglobin (Hb) lower than 6 g/dl. On April 13, he suddenly had a high fever. Under empirical anti-infection, his high fever dropped and maintained at a low fever level; however, his anemia worsened. On April 25, he was transferred to the medical intensive care unit (MICU) due to severe pneumonia, acute respiratory distress syndrome (ARDS), acute aplastic crisis (AAC), and hemophagocytic syndrome (HPS), which were subsequently confirmed to be related to B19 infection. After methylprednisolone, intravenous immunoglobulin (IVIG), empirical anti-infection, and supportive treatment, the lung infection improved, but hematopoietic and liver abnormalities aggravated, and systemic B19 infection occurred. Finally, the patient developed a refractory arrhythmia, heart failure, and shock and was referred to a local hospital by his family on May 8, 2021. Unfortunately, he died the next day. Fourteen days after he was transferred to MICU, seven nurses caring for him in his first two days in the MICU developed self-limiting erythema infectiosum (EI). Conclusions: B19 infection is self-limiting in healthy people, with low virulence and infectivity; however, in AH patients with HA, it can lead to fatal consequences and high contagion.


Assuntos
Anemia Hemolítica/imunologia , Hepatite Alcoólica/imunologia , Insuficiência de Múltiplos Órgãos/imunologia , Infecções por Parvoviridae/imunologia , Parvovirus B19 Humano/imunologia , Adulto , Hepatite Alcoólica/diagnóstico , Humanos , Masculino , Insuficiência de Múltiplos Órgãos/diagnóstico , Infecções por Parvoviridae/diagnóstico
4.
Cells ; 10(11)2021 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-34831101

RESUMO

As the number of confirmed cases and deaths occurring from Coronavirus disease 2019 (COVID-19) surges worldwide, health experts are striving hard to fully comprehend the extent of damage caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Although COVID-19 primarily manifests itself in the form of severe respiratory distress, it is also known to cause systemic damage to almost all major organs and organ systems within the body. In this review, we discuss the molecular mechanisms leading to multi-organ failure seen in COVID-19 patients. We also examine the potential of stem cell therapy in treating COVID-19 multi-organ failure cases.


Assuntos
COVID-19/complicações , COVID-19/terapia , Insuficiência de Múltiplos Órgãos/etiologia , Insuficiência de Múltiplos Órgãos/terapia , Transplante de Células-Tronco , COVID-19/imunologia , Ensaios Clínicos como Assunto , Síndrome da Liberação de Citocina/etiologia , Síndrome da Liberação de Citocina/imunologia , Síndrome da Liberação de Citocina/terapia , Humanos , Imunomodulação , Insuficiência de Múltiplos Órgãos/imunologia , Medicina Regenerativa , SARS-CoV-2/patogenicidade , Células-Tronco/citologia , Células-Tronco/imunologia
5.
Immunopharmacol Immunotoxicol ; 43(6): 633-643, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34647511

RESUMO

The coronavirus disease-19 (COVID-19), at first, was reported in Wuhan, China, and then rapidly became pandemic throughout the world. Cytokine storm syndrome (CSS) in COVID-19 patients is associated with high levels of cytokines and chemokines that cause multiple organ failure, systemic inflammation, and hemodynamic instabilities. Acute respiratory distress syndrome (ARDS), a common complication of COVID-19, is a consequence of cytokine storm. In this regard, several drugs have been being investigated to suppress this inflammatory condition. Purinergic signaling receptors comprising of P1 adenosine and P2 purinoceptors play a critical role in inflammation. Therefore, activation or inhibition of some subtypes of these kinds of receptors is most likely to be beneficial to attenuate cytokine storm. This article summarizes suggested therapeutic drugs with potential anti-inflammatory effects through purinergic receptors.


Assuntos
Anti-Inflamatórios/uso terapêutico , Tratamento Farmacológico da COVID-19 , Síndrome da Liberação de Citocina/prevenção & controle , Citocinas/sangue , Antagonistas Purinérgicos/uso terapêutico , Receptores Purinérgicos/efeitos dos fármacos , SARS-CoV-2/efeitos dos fármacos , Animais , Anti-Inflamatórios/efeitos adversos , Biomarcadores/sangue , COVID-19/sangue , COVID-19/imunologia , COVID-19/virologia , Síndrome da Liberação de Citocina/sangue , Síndrome da Liberação de Citocina/imunologia , Síndrome da Liberação de Citocina/virologia , Interações Hospedeiro-Patógeno , Humanos , Ligantes , Terapia de Alvo Molecular , Insuficiência de Múltiplos Órgãos/imunologia , Insuficiência de Múltiplos Órgãos/prevenção & controle , Insuficiência de Múltiplos Órgãos/virologia , Antagonistas Purinérgicos/efeitos adversos , Receptores Purinérgicos/metabolismo , SARS-CoV-2/imunologia , SARS-CoV-2/patogenicidade , Transdução de Sinais
6.
Eur J Pharmacol ; 906: 174279, 2021 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-34197778

RESUMO

Acute pancreatitis (AP) is a common pancreatic inflammation associated with substantial morbidity and mortality. AP may be mild or severe which can spread systemically causing multiple organs failure (MOF) and even death. In the current study, protocatechuic acid (PCA), a natural phenolic acid, was investigated for its possible protective potential against L-arginine induced AP and multiple organs injury (MOI) in rats. AP was induced by L-arginine (500 mg/100 g, ip). Two dose levels of PCA were tested (50 and 100 mg/kg, oral, 10 days before L-arginine injection). PCA successfully protected against L-arginine induced AP and MOI that was manifested by normalizing pancreatic, hepatic, pulmonary, and renal tissue architecture and restoring the normal values of pancreatic enzymes (amylase and lipase), serum total protein, liver enzymes (alanine transaminase (ALT) and aspartate transaminase (AST)) and kidney function biomarkers (blood urea nitrogen (BUN) and serum creatinine (Cr)) that were significantly elevated upon L-arginine administration. Additionally, PCA restored balanced oxidant/antioxidants status that was disrupted by L-arginine and normalized pancreatic levels of inducible nitric oxide synthase (iNOS) and nitric oxide (NO) content. Moreover, PCA significantly decreased L-arginine induced elevation in pancreatic high motility group box protein 1 (HMGB1), toll like receptor 4 (TLR4), myeloid differentiation factor 88 (MyD88), nuclear factor kappa B (NF-κB), tumor necrosis factor- α (TNF-α), interleukin-1ß (IL-1ß), and interleukin-6 (IL-6) expression. PCA significantly ameliorated L-arginine-induced AP and MOI through its anti-inflammatory and antioxidant effects. HMGB1/TLR4/NF-κB was the major pathway involved in the observed protective potential.


Assuntos
Anti-Inflamatórios/farmacologia , Hidroxibenzoatos/farmacologia , Insuficiência de Múltiplos Órgãos/prevenção & controle , Pancreatite/prevenção & controle , Animais , Anti-Inflamatórios/uso terapêutico , Arginina/administração & dosagem , Arginina/toxicidade , Modelos Animais de Doenças , Proteína HMGB1/metabolismo , Humanos , Hidroxibenzoatos/uso terapêutico , Rim/efeitos dos fármacos , Rim/imunologia , Rim/patologia , Fígado/efeitos dos fármacos , Fígado/imunologia , Fígado/patologia , Masculino , Insuficiência de Múltiplos Órgãos/induzido quimicamente , Insuficiência de Múltiplos Órgãos/imunologia , Insuficiência de Múltiplos Órgãos/patologia , NF-kappa B/metabolismo , Pâncreas/efeitos dos fármacos , Pâncreas/imunologia , Pâncreas/patologia , Pancreatite/induzido quimicamente , Pancreatite/imunologia , Pancreatite/patologia , Ratos , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/metabolismo
7.
J Trauma Acute Care Surg ; 91(4): 700-707, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34238858

RESUMO

BACKGROUND: Inflammatory lipid mediators in mesenteric lymph (ML), including arachidonic acid (AA), are considered to play an important role in the pathogenesis of multiple-organ dysfunction after hemorrhagic shock. A previous study suggested that vagus nerve stimulation (VNS) could relieve shock-induced gut injury and abrogate ML toxicity, resulting in the prevention of multiple-organ dysfunction. However, the detailed mechanism of VNS in lymph toxicity remains unclear. The study aimed to investigate the relationship between VNS and inflammatory lipid mediators in ML. METHODS: Male Sprague-Dawley rats underwent laparotomy and superior mesenteric artery obstruction (SMAO) for 60 minutes to induce intestinal ischemia followed by reperfusion and observation. The ML duct was cannulated, and ML samples were obtained both before and after SMAO. The distal ileum was removed at the end of the observation period. In one group of animals, VNS was performed from 10 minutes before 10 minutes after SMAO (5 V, 0.5 Hz). Liquid chromatography-electrospray ionization-tandem mass spectrometry analysis of AA was performed for each ML sample. The biological activity of ML was examined using a monocyte nuclear factor κ-light-chain-enhancer of activated B cells activation assay. Western blotting of phospholipase A2 group IIA (PLA2-IIA) was also performed for ML and ileum samples. RESULTS: Vagus nerve stimulation relieved the SMAO-induced histological gut injury. The concentration of AA and level of nuclear factor κ-light-chain-enhancer of activated B cells activation in ML increased significantly after SMAO, whereas VNS prevented these responses. Western blotting showed PLA2-IIA expression in the ML and ileum after SMAO; however, the appearance of PLA2-IIA band was remarkably decreased in the samples from VNS-treated animals. CONCLUSION: The results suggested that VNS could relieve gut injury induced by SMAO and decrease the production of AA in ML by altering PLA2-IIA expression in the gut and ML.


Assuntos
Ácido Araquidônico/metabolismo , Insuficiência de Múltiplos Órgãos/prevenção & controle , Traumatismo por Reperfusão/terapia , Choque Hemorrágico/complicações , Estimulação do Nervo Vago , Animais , Modelos Animais de Doenças , Humanos , Linfa/imunologia , Linfa/metabolismo , Vasos Linfáticos/patologia , Masculino , Mesentério/patologia , Insuficiência de Múltiplos Órgãos/imunologia , Insuficiência de Múltiplos Órgãos/patologia , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/imunologia , Traumatismo por Reperfusão/patologia , Choque Hemorrágico/imunologia
8.
Pharmacol Res ; 172: 105781, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34302975

RESUMO

Sepsis is a severe inflammatory disorder that can lead to multiple organ injury. Isosteviol sodium (STV-Na) is a terpenoid derived from stevioside that exerts anti-inflammatory, antioxidant and antiapoptotic activities. However, the influence of STV-Na on sepsis remains unknown. Here, we assessed the potential effects of STV-Na on sepsis and multiple organ injury induced by lipopolysaccharide (LPS). We found that STV-Na increased the survival rate of mice treat with LPS, significantly improved the functions of the heart, lung, liver, and kidney, reduced the production of inflammatory cytokines and decreased macrophage infiltration. Moreover, Multiorgan metabolomics analysis demonstrated that glutathione metabolism, purine metabolism, glycerophospholipid metabolism and pantothenate and CoA biosynthesis, were significantly altered by STV-Na. This study provides novel insights into the metabolite changes of multiple organ injury in septic mice, which may help characterize the underlying mechanism and provide an improved understanding of the therapeutic effects of STV-Na on sepsis.


Assuntos
Anti-Inflamatórios/uso terapêutico , Diterpenos do Tipo Caurano/uso terapêutico , Insuficiência de Múltiplos Órgãos/tratamento farmacológico , Sepse/tratamento farmacológico , Animais , Anti-Inflamatórios/farmacologia , Diterpenos do Tipo Caurano/farmacologia , Glutationa/metabolismo , Glicerofosfolipídeos/metabolismo , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Lipopolissacarídeos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Masculino , Metabolômica , Camundongos Endogâmicos BALB C , Insuficiência de Múltiplos Órgãos/etiologia , Insuficiência de Múltiplos Órgãos/imunologia , Insuficiência de Múltiplos Órgãos/metabolismo , Miocárdio/metabolismo , Miocárdio/patologia , Ácido Pantotênico/metabolismo , Purinas/metabolismo , Sepse/complicações , Sepse/imunologia , Sepse/metabolismo , Baço/efeitos dos fármacos , Baço/metabolismo , Baço/patologia
9.
Blood ; 138(2): 178-189, 2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-33598692

RESUMO

Activation of coagulation factor (F) XI promotes multiorgan failure in rodent models of sepsis and in a baboon model of lethal systemic inflammation induced by infusion of heat-inactivated Staphylococcus aureus. Here we used the anticoagulant FXII-neutralizing antibody 5C12 to verify the mechanistic role of FXII in this baboon model. Compared with untreated control animals, repeated 5C12 administration before and at 8 and 24 hours after bacterial challenge prevented the dramatic increase in circulating complexes of contact system enzymes FXIIa, FXIa, and kallikrein with antithrombin or C1 inhibitor, and prevented cleavage and consumption of high-molecular-weight kininogen. Activation of several coagulation factors and fibrinolytic enzymes was also prevented. D-dimer levels exhibited a profound increase in the untreated animals but not in the treated animals. The antibody also blocked the increase in plasma biomarkers of inflammation and cell damage, including tumor necrosis factor, interleukin (IL)-1ß, IL-6, IL-8, IL-10, granulocyte-macrophage colony-stimulating factor, nucleosomes, and myeloperoxidase. Based on clinical presentation and circulating biomarkers, inhibition of FXII prevented fever, terminal hypotension, respiratory distress, and multiorgan failure. All animals receiving 5C12 had milder and transient clinical symptoms and were asymptomatic at day 7, whereas untreated control animals suffered irreversible multiorgan failure and had to be euthanized within 2 days after the bacterial challenge. This study confirms and extends our previous finding that at least 2 enzymes of the contact activation complex, FXIa and FXIIa, play critical roles in the development of an acute and terminal inflammatory response in baboons challenged with heat-inactivated S aureus.


Assuntos
Fator XII/metabolismo , Insuficiência de Múltiplos Órgãos/metabolismo , Insuficiência de Múltiplos Órgãos/microbiologia , Staphylococcus aureus/fisiologia , Animais , Anticorpos/uso terapêutico , Transtornos da Coagulação Sanguínea/complicações , Transtornos da Coagulação Sanguínea/imunologia , Transtornos da Coagulação Sanguínea/microbiologia , Plaquetas/metabolismo , Microambiente Celular , Ativação do Complemento , Fator XII/imunologia , Feminino , Fibrinogênio/metabolismo , Temperatura Alta , Inflamação/complicações , Inflamação/patologia , Masculino , Insuficiência de Múltiplos Órgãos/imunologia , Papio , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/patologia , Análise de Sobrevida
11.
Am J Respir Crit Care Med ; 203(2): 192-201, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33217246

RESUMO

Rationale: In life-threatening coronavirus disease (COVID-19), corticosteroids reduce mortality, suggesting that immune responses have a causal role in death. Whether this deleterious inflammation is primarily a direct reaction to the presence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or an independent immunopathologic process is unknown.Objectives: To determine SARS-CoV-2 organotropism and organ-specific inflammatory responses and the relationships among viral presence, inflammation, and organ injury.Methods: Tissue was acquired from 11 detailed postmortem examinations. SARS-CoV-2 organotropism was mapped by using multiplex PCR and sequencing, with cellular resolution achieved by in situ viral S (spike) protein detection. Histologic evidence of inflammation was quantified from 37 anatomic sites, and the pulmonary immune response was characterized by using multiplex immunofluorescence.Measurements and Main Results: Multiple aberrant immune responses in fatal COVID-19 were found, principally involving the lung and reticuloendothelial system, and these were not clearly topologically associated with the virus. Inflammation and organ dysfunction did not map to the tissue and cellular distribution of SARS-CoV-2 RNA and protein between or within tissues. An arteritis was identified in the lung, which was further characterized as a monocyte/myeloid-rich vasculitis, and occurred together with an influx of macrophage/monocyte-lineage cells into the pulmonary parenchyma. In addition, stereotyped abnormal reticuloendothelial responses, including excessive reactive plasmacytosis and iron-laden macrophages, were present and dissociated from viral presence in lymphoid tissues.Conclusions: Tissue-specific immunopathology occurs in COVID-19, implicating a significant component of the immune-mediated, virus-independent immunopathologic process as a primary mechanism in severe disease. Our data highlight novel immunopathologic mechanisms and validate ongoing and future efforts to therapeutically target aberrant macrophage and plasma-cell responses as well as promote pathogen tolerance in COVID-19.


Assuntos
COVID-19/imunologia , Inflamação/virologia , Pulmão/imunologia , Insuficiência de Múltiplos Órgãos/virologia , SARS-CoV-2/imunologia , Idoso , Idoso de 80 Anos ou mais , Autopsia , Biópsia , COVID-19/patologia , COVID-19/virologia , Teste de Ácido Nucleico para COVID-19 , Feminino , Imunofluorescência , Humanos , Inflamação/imunologia , Inflamação/patologia , Pulmão/patologia , Pulmão/virologia , Masculino , Insuficiência de Múltiplos Órgãos/imunologia , Insuficiência de Múltiplos Órgãos/patologia , SARS-CoV-2/patogenicidade , Índice de Gravidade de Doença
12.
J Immunol ; 205(6): 1633-1643, 2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32769121

RESUMO

The inability to effectively control invading bacteria or other pathogens is a major cause of multiple organ dysfunction and death in sepsis. As the first-line defense of the immune system, macrophages play a crucial role in the removal of pathogens during sepsis. In this study, we define secreted and transmembrane 1A (Sectm1a) as a novel ligand of glucocorticoid-induced TNFR (GITR) that greatly boosts macrophage phagocytosis and bactericidal capacity. Using a global Sectm1a knockout (KO) mouse model, we observed that Sectm1a deficiency significantly suppressed phagocytosis and bactericidal activity in both recruited macrophages and tissue-resident macrophages, which consequently aggravated bacterial burden in the blood and multiple organs and further increased systemic inflammation, leading to multiple organ injury and increased mortality during polymicrobial sepsis. By contrast, treatment of septic mice with recombinant Sectm1a protein (rSectm1a) not only promoted macrophage phagocytosis and bactericidal activity but also significantly improved survival outcome. Mechanistically, we identified that Sectm1a could bind to GITR in the surface of macrophages and thereby activate its downstream PI3K-Akt pathway. Accordingly, rSectm1a-mediated phagocytosis and bacterial killing were abolished in macrophages by either KO of GITR or pharmacological inhibition of the PI3K-Akt pathway. In addition, rSectm1a-induced therapeutic effects on sepsis injury were negated in GITR KO mice. Taken together, these results uncover that Sectm1a may represent a novel target for drug development to control bacterial dissemination during sepsis or other infectious diseases.


Assuntos
Proteína Relacionada a TNFR Induzida por Glucocorticoide/metabolismo , Macrófagos/fisiologia , Proteínas de Membrana/metabolismo , Insuficiência de Múltiplos Órgãos/imunologia , Sepse/imunologia , Animais , Proteína Relacionada a TNFR Induzida por Glucocorticoide/genética , Humanos , Tolerância Imunológica , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Oncogênica v-akt/metabolismo , Fagocitose , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais
14.
Int Immunopharmacol ; 78: 106049, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31830624

RESUMO

AIMS: Sepsis is defined as a life-threatening organ dysfunction that is caused by a dysregulated host response to infection. Although much progress has been made in understanding the pathophysiology of sepsis, further discussion and study of the detailed therapeutic mechanisms are needed. Autophagy and endoplasmic reticulum stress are two pathways of the complicated regulatory network of sepsis. Herein, we focus on the cellular mechanism in which autophagy and endoplasmic reticulum stress participate in hydrogen (H2)-protected sepsis-induced organ injury. MATERIALS AND METHODS: Male C57BL/6 mice were randomly divided into the following groups: control group, cecal ligation puncture (CLP) group, CLP + tunicamycin(TM) group, CLP + 4-phenyl butyric acid (4-PBA) group, CLP + rapamycin (Rap) group, CLP + 3-methyladenine (3-MA) group, CLP + H2 group, CLP + H2 + 3-MA group, and CLP + H2 + TM group. After the experiment was completed, autophagosome was detected by transmission electron microscopy; protein PKR-like ER kinase (PERK), p-PERK, Eukaryotic translation initiation factor-2α (eIF2α), p-eIF2α, inositol-requiring enzyme1α(IRE1α), C/EBP homologous protein(CHOP), activating transcription factor(ATF), XBP-1, microtubule-associated protein 1 light(LC3), Beclin1, PTEN-induced putative kinase 1(PINK1), Parkin, and p65 subunit of Nuclear factor kappa B(NF-κb) were measured by Western blot; myeloperoxidase(MPO) activity in lung, bronchoalveolar lavage(BAL) total protein, lung wet-to-dry(W/D) ratio, serum biochemical indicators, 7-day survival rate, and pathological injury scores of lung, liver, and kidney were tested; and cytokines tumor necrosis factor-α(TNF-α), Interleukin(IL)-1ß, and IL-6 and high mobility group box protein (HMGB)1 were detected by enzyme-linked immunosorbent assay(ELISA). RESULTS: We demonstrated that sepsis induced endoplasmic reticulum stress. Moreover, it was found that an increase in endoplasmic reticulum impaired autophagy activity in sepsis, and the absence of endoplasmic reticulum stress attenuated tissue histological injury and dysfunction of lung, liver, and kidney in septic mice. Intriguingly, hydrogen alleviated the endoplasmic reticulum stress via the autophagy pathway and also mitigated inflammation and organ injury. CONCLUSION: Hydrogen provided protection from organ injury induced by sepsis via autophagy activation and endoplasmic reticulum stress pathway inactivation.


Assuntos
Autofagia/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Hidrogênio/administração & dosagem , Insuficiência de Múltiplos Órgãos/prevenção & controle , Sepse/tratamento farmacológico , Animais , Autofagia/imunologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Estresse do Retículo Endoplasmático/imunologia , Humanos , Hidrogênio/química , Injeções Intraperitoneais , Masculino , Camundongos , Insuficiência de Múltiplos Órgãos/imunologia , Solução Salina/administração & dosagem , Solução Salina/química , Sepse/complicações , Sepse/imunologia
15.
Int J Mol Sci ; 20(23)2019 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-31801287

RESUMO

Multiple organ dysfunction syndrome (MODS) caused by the systemic inflammatory response during sepsis is responsible for millions of deaths worldwide each year, and despite broad consensus concerning its pathophysiology, no specific or effective therapies exist. Recent efforts to treat and/or prevent MODS have included a variety of biologics, recombinant proteins targeting various components of the host response to the infection (e.g., inflammation, coagulation, etc.) Improvements in molecular biology and pharmaceutical engineering have enabled a wide range of utility for biologics to target various aspects of the systemic inflammatory response. The majority of clinical trials to date have failed to show clinical benefit, but some have demonstrated promising results in certain patient populations. In this review we summarize the underlying rationale and outcome of major clinical trials where biologics have been tested as a pharmacotherapy for MODS in sepsis. A brief description of the study design and overall outcome for each of the major trials are presented. Emphasis is placed on discussing targets and/or trials where promising results were observed. Post hoc analyses of trials where therapy demonstrated harm or additional risk to certain patient subgroups are highlighted, and details are provided about specific trials where more stringent inclusion/exclusion criteria are warranted.


Assuntos
Anti-Inflamatórios/uso terapêutico , Produtos Biológicos/uso terapêutico , Coagulação Sanguínea/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Insuficiência de Múltiplos Órgãos/tratamento farmacológico , Sepse/tratamento farmacológico , 1-Alquil-2-acetilglicerofosfocolina Esterase/uso terapêutico , Fosfatase Alcalina/uso terapêutico , Coagulação Sanguínea/genética , Coagulação Sanguínea/imunologia , Ensaios Clínicos como Assunto , Citocinas/antagonistas & inibidores , Citocinas/genética , Citocinas/imunologia , Expressão Gênica , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Humanos , Imunoglobulinas Intravenosas/uso terapêutico , Inflamação , Insuficiência de Múltiplos Órgãos/genética , Insuficiência de Múltiplos Órgãos/imunologia , Insuficiência de Múltiplos Órgãos/patologia , Sepse/genética , Sepse/imunologia , Sepse/patologia
16.
Cytometry A ; 95(11): 1167-1177, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31595661

RESUMO

Severe injury and hemorrhagic shock (HS) result in multiple changes to hematopoietic differentiation, which contribute to the development of immunosuppression and multiple organ failure (MOF). Understanding the changes that take place during the acute injury phase may help predict which patients will develop MOF and provide potential targets for therapy. Obtaining bone marrow from humans during the acute injury phase is difficult so published data are largely derived from peripheral blood samples, which infer bone marrow changes that reflect the sustained inflammatory response. This preliminary and opportunistic study investigated leucopoietic changes in rat bone marrow 6 h following traumatic injury and HS. Terminally anesthetized male Porton Wistar rats were allocated randomly to receive a sham operation (cannulation with no injury) or femoral fracture and HS. Bone marrow cells were flushed from rat femurs and immunophenotypically stained with specific antibody panels for lymphoid (CD45R, CD127, CD90, and IgM) or myeloid (CD11b, CD45, and RP-1) lineages. Subsequently, cell populations were fluorescence-activated cell sorted for morphological assessment. Stage-specific cell populations were identified using a limited number of antibodies, and leucopoietic changes were determined 6 h following trauma and HS. Myeloid subpopulations could be identified by varying levels CD11b expression, CD45, and RP-1. Trauma and HS resulted in a significant reduction in total CD11b + myeloid cells including both immature (RP-1(-)) and mature (RP-1+) granulocytes. Multiple B-cell lymphoid subsets were identified. The total percentage of CD90+ subsets remained unchanged following trauma and HS, but there was a reduction in the numbers of maturing CD90(-) cells suggesting movement into the periphery. © 2019 The Authors. Cytometry Part A published by Wiley Periodicals, Inc. on behalf of International Society for Advancement of Cytometry.


Assuntos
Células da Medula Óssea/citologia , Fraturas do Fêmur/imunologia , Células-Tronco Hematopoéticas/citologia , Choque Hemorrágico/imunologia , Ferimentos e Lesões/imunologia , Animais , Peptídeos Catiônicos Antimicrobianos/metabolismo , Linfócitos B/citologia , Linfócitos B/metabolismo , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Antígeno CD11b/metabolismo , Linhagem da Célula/imunologia , Citometria de Fluxo , Granulócitos/citologia , Granulócitos/metabolismo , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/metabolismo , Imunofenotipagem , Inflamação/imunologia , Inflamação/metabolismo , Antígenos Comuns de Leucócito/metabolismo , Linfopoese/imunologia , Masculino , Insuficiência de Múltiplos Órgãos/imunologia , Insuficiência de Múltiplos Órgãos/patologia , Células Mieloides/citologia , Células Mieloides/metabolismo , Ratos , Ratos Wistar , Choque Hemorrágico/metabolismo , Antígenos Thy-1/metabolismo , Ferimentos e Lesões/metabolismo
17.
Mediators Inflamm ; 2019: 6197084, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30881224

RESUMO

Sepsis remains one of the leading causes of mortality in intensive care units, but there is a shortage of effective treatments. A dysregulated host immune response and multiple organ injury are major factors for the pathogenesis and progression of sepsis, which require specific mechanism and treatment. In the present study, we performed an intracerebroventricular (ICV) injection of BoxA, a specific antagonist of high-mobility group box 1 protein (HMGB1), in septic rats that were produced by cecal ligation and puncture surgery; we further assessed the functional changes of multiple organs and splenic T lymphocytes. We found that the inhibition of cerebral HMGB1 significantly alleviated multiple organ damage under septic exposure, including damage to the heart, liver, lungs, and kidneys; reversed the immune dysfunction of T cells; and increased the survival of septic rats. These data suggest that central HMGB1 might be a potential therapeutic target for septic challenge and that inhibition of brain HMGB1 can protect against multiple organ dysfunction induced by sepsis.


Assuntos
Proteína HMGB1/metabolismo , Imunidade Celular/imunologia , Insuficiência de Múltiplos Órgãos/metabolismo , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Proteína HMGB1/antagonistas & inibidores , Infusões Intraventriculares , Masculino , Insuficiência de Múltiplos Órgãos/imunologia , Ratos , Ratos Sprague-Dawley , Sepse/imunologia , Sepse/metabolismo
18.
Anaesth Crit Care Pain Med ; 38(5): 485-491, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30797048

RESUMO

BACKGROUND: Sepsis profoundly alters immune homeostasis. Cytokine release after whole blood lipopolysaccharide (LPS)-stimulation reflects cell function across multiple immune cell classes and represents the immune response to LPS. The main goal of this study was to evaluate the prognostic value of ex vivo stimulation of whole blood with LPS in sepsis. METHODS: Blood was drawn on day 1 and day 7 after admission, and stimulated ex vivo with LPS. Tumour necrosis factor (TNF)-α, interleukin (IL)-1ß, IL-6 and IL-10 were measured with and without stimulation. Our primary outcome measure was the persistence of at least one organ dysfunction at day 7. Organ dysfunction was defined according to the SOFA components by a score ≥ 2. RESULTS: Forty-nine patients with sepsis from a 21-bed intensive care unit, and 23 healthy volunteers were enrolled. The blood of septic patients was less responsive to ex vivo stimulation with LPS than that of healthy controls at day 1 and 7, as demonstrated by lower TNF-α, IL-1ß, IL-6 and IL-10 release. Persistent organ dysfunction was more frequent in patients with lower IL-10 release at day 1 but such an association was not found for pro-inflammatory cytokines. A persistent low IL-10 release at day 7 was also associated with persistent organ dysfunction. CONCLUSION: These data suggest that the capacity to produce IL-10 in response to whole blood ex vivo stimulation early in sepsis, as well as persistent low IL-10 response over time, may help in prognostication and patient stratification. These results will need to be confirmed in future studies.


Assuntos
Interleucina-10/sangue , Interleucina-1beta/sangue , Interleucina-6/sangue , Insuficiência de Múltiplos Órgãos/sangue , Sepse/sangue , Fator de Necrose Tumoral alfa/sangue , Idoso , Estudos de Casos e Controles , Feminino , Humanos , Lipopolissacarídeos , Masculino , Pessoa de Meia-Idade , Insuficiência de Múltiplos Órgãos/imunologia , Escores de Disfunção Orgânica , Avaliação de Resultados em Cuidados de Saúde , Prognóstico , Estudos Prospectivos , Sepse/imunologia , Fatores de Tempo
19.
Kidney Int ; 95(4): 859-879, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30777286

RESUMO

Intestinal Paneth cells play a critical role in ischemic acute kidney injury (AKI) by releasing interleukin 17A (IL-17A). Because Toll-like receptor 9 (TLR9) activation degranulates Paneth cells and necrotic tubular epithelial cells release several damage associated molecular patterns that target TLR9, we tested the hypothesis that intestinal TLR9 deficiency would protect against ischemic AKI and associated remote intestinal and hepatic dysfunction by decreasing Paneth cell degranulation. We generated mice lacking TLR9 in intestinal epithelia (TLR9fl/fl Villin Cre mice) and compared them to wild type (TLR9fl/fl) mice following right nephrectomy and left ischemia/reperfusion. To our surprise, mice lacking intestinal TLR9 had exacerbated kidney, liver, and small intestine injury after ischemia/reperfusion compared to wild type mice, characterized by increased kidney and intestinal inflammation, apoptosis, and necrosis as well as increased hepatic inflammation and apoptosis. Mice lacking intestinal TLR9 had larger Paneth cell granule size, pronounced intestinal macrophage infiltration, and higher intestinal crypt IL-17A expression. Administration of IL-17A neutralizing antibody prevented the exacerbation of ischemic AKI in mice lacking intestinal TLR9. These studies suggest that intestinal TLR9 activation protects against ischemic AKI and associated remote multi-organ dysfunction syndrome by regulating Paneth cell IL-17A synthesis.


Assuntos
Injúria Renal Aguda/imunologia , Interleucina-17/metabolismo , Insuficiência de Múltiplos Órgãos/imunologia , Celulas de Paneth/patologia , Receptor Toll-Like 9/metabolismo , Injúria Renal Aguda/patologia , Animais , Apoptose , Modelos Animais de Doenças , Progressão da Doença , Humanos , Hiperplasia/imunologia , Hiperplasia/patologia , Interleucina-17/antagonistas & inibidores , Interleucina-17/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Intestinos/imunologia , Intestinos/patologia , Rim/imunologia , Rim/patologia , Fígado/imunologia , Fígado/patologia , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Transgênicos , Insuficiência de Múltiplos Órgãos/patologia , Celulas de Paneth/imunologia , Celulas de Paneth/metabolismo , Traumatismo por Reperfusão/imunologia , Traumatismo por Reperfusão/patologia , Receptor Toll-Like 9/genética
20.
Eur Rev Med Pharmacol Sci ; 23(2): 771-787, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30720186

RESUMO

Acute pancreatitis (AP) is the most common gastrointestinal disorder requiring hospitalization, with a high rate of morbidity and mortality. Severe AP is characterized by the presence of persistent organ failure involving single or multiple organs. Clinical evolution, laboratory and radiological assessment are necessary to evaluate the prognosis and inform the management of AP. The onset of severe AP may be classified in two principal phases. The early phase, during the first week, is characterized by the activation of the auto-inflammatory cascade, gut dysbiosis, bacterial translocation, and the down-regulation of immune responses. The late phase is characterized by the development of local and systemic complications. Several old paradigms have been amended in the management of AP patients, such as the indication of nutrition, the use of antibiotic therapy, pain control strategies, and even the use of surgery. Real world evidence has shown that in the majority of cases a step-up approach is most effective. In this review, we discuss the clinical assessment and improvements to the management of patients with severe AP in a high volume center where a multi-disciplinary approach is performed.


Assuntos
Insuficiência de Múltiplos Órgãos/terapia , Dor/tratamento farmacológico , Pancreatite/terapia , Equipe de Assistência ao Paciente , Analgésicos/uso terapêutico , Antibacterianos/uso terapêutico , Translocação Bacteriana/imunologia , Colangiopancreatografia Retrógrada Endoscópica , Drenagem/métodos , Gastroenterostomia , Microbioma Gastrointestinal/imunologia , Humanos , Insuficiência de Múltiplos Órgãos/imunologia , Terapia Nutricional/métodos , Dor/imunologia , Manejo da Dor/métodos , Pâncreas/diagnóstico por imagem , Pâncreas/imunologia , Pâncreas/patologia , Pâncreas/cirurgia , Pancreatite/complicações , Pancreatite/diagnóstico , Pancreatite/imunologia , Índice de Gravidade de Doença , Tomografia Computadorizada por Raios X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA