Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
1.
Curr Gene Ther ; 24(5): 410-421, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39005063

RESUMO

BACKGROUND: Injury systemically disrupts the homeostatic balance and can cause organ failure. LF mediates both iron-dependent and iron-independent mechanisms, and the role of LF in regulating iron homeostasis is vital in terms of metabolism. OBJECTIVES: In this study, we evaluated the organ-level effect and gene expression change of bLf in the cutaneous repair process. MATERIALS AND METHODS: An excisional full-thickness skin defect (FTSD) wound model was created in male Sprague Dawley rats (180-250 g) (n = 48) fed a high-fat diet (HFD) and the PHGPx, SLC7A11 and SLC40A1 genes and iron metabolism were evaluated. The animals were randomly divided into 6 groups: 1- Control, 2- bLf (200 mg/kg/day, oral), 3- FTSD (12 mm in diameter, dorsal), 4- HFD + bLf, 5- HFD + FTSD, 6- HFD + FTSD + bLf. Histologically, iron accumulation was demonstrated by Prussian blue staining in the liver, kidney, and intestinal tissues. Gene expression analysis was performed with qPCR. RESULTS: Histologically, iron accumulation was demonstrated by Prussian blue staining in the liver, kidney, and intestinal tissues. Prussian blue reactions were detected in the kidney. PHPGx and SLC7A11 genes in kidney and liver tissue were statistically significant (P < 0.05) except for the SLC40A1 gene (P > 0.05). Expression changes of the three genes were not statistically significant in analyses of rat intestinal tissue (P = 0.057). CONCLUSION: In the organ-level ferroptotic damage mechanism triggered by wound formation. BLf controls the expression of three genes and manages iron deposition in these three tissues. In addition, it suppressed the increase in iron that would drive the cell to ferroptosis and anemia caused by inflammation, thereby eliminating iron deposition in the tissues.


Assuntos
Homeostase , Ferro , Lactoferrina , Ratos Sprague-Dawley , Cicatrização , Animais , Ferro/metabolismo , Ratos , Masculino , Homeostase/efeitos dos fármacos , Lactoferrina/farmacologia , Lactoferrina/genética , Cicatrização/efeitos dos fármacos , Cicatrização/genética , Bovinos , Insuficiência de Múltiplos Órgãos/genética , Insuficiência de Múltiplos Órgãos/metabolismo , Insuficiência de Múltiplos Órgãos/tratamento farmacológico , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Fígado/metabolismo , Fígado/efeitos dos fármacos
2.
Immunol Invest ; 52(5): 583-597, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37216493

RESUMO

Systemic inflammatory response syndrome (SIRS) frequently accompanies early postoperative period after cardiac surgery and in some cases is complicated by multiple organ failure (MOF). Inherited variation in the innate immune response genes (e.g., TREM1) is among the major factors determining the development of SIRS and the risk of MOF. This research was aimed to study whether the polymorphisms within the TREM1 gene are associated with MOF after the coronary artery bypass graft (CABG) surgery. Here we enrolled 592 patients who underwent CABG surgery in the Research Institute for Complex Issues of Cardiovascular Diseases (Kemerovo, Russia) and documented 28 cases of MOF. Genotyping was performed by allele-specific PCR using TaqMan probes. In addition, we measured serum soluble triggering receptor expressed on myeloid cells 1 (sTREM-1) using enzyme-linked immunosorbent assay. Five polymorphisms (rs1817537, rs2234246, rs3804277, rs7768162 andrs4711668) within the TREM1 gene were significantly associated with MOF. Patients with MOF had higher serum sTREM-1 as compared with those without MOF at both pre- and post-intervention stages. Serum sTREM-1 was associated with the rs1817537,rs2234246 and rs3804277 polymorphisms within the TREM1 gene. Minor alleles within the TREM1 gene define the level of serum sTREM-1 and are associated with MOF after CABG surgery.


Assuntos
Procedimentos Cirúrgicos Cardíacos , Glicoproteínas de Membrana , Humanos , Receptor Gatilho 1 Expresso em Células Mieloides/genética , Glicoproteínas de Membrana/genética , Receptores Imunológicos/genética , Insuficiência de Múltiplos Órgãos/genética , Síndrome de Resposta Inflamatória Sistêmica , Procedimentos Cirúrgicos Cardíacos/efeitos adversos , Biomarcadores
3.
Int J Mol Sci ; 23(23)2022 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-36499077

RESUMO

Assess the level of mitochondrial DNA depending on the presence of multiple organ failure in patients after heart surgery. The study included 60 patients who underwent surgical treatment of valvular heart disease using cardiopulmonary bypass. Uncomplicated patients were included in the 1st group (n = 30), patients with complications and multiple organ failure (MOF) were included in the 2nd group (n = 30). Serum mtDNA levels were determined by quantitative real-time polymerase chain reaction with fluorescent dyes. Mitochondrial DNA gene expression did not differ between group before surgery. Immediately after the intervention, cytochrome B gene expression was higher in the group with MOF, and it remained high during entire follow-up period. A similar trend was observed in cytochrome oxidase gene expression. Increased NADH levels of gene expressions during the first postoperative day were noted in both groups, the expression showed tendency to increase on the third postoperative day. mtDNA gene expression in the "MOF present" group remained at a higher level compared with the group without complications. A positive correlation was reveled between the severity of MOF according to SOFA score and the level of mtDNA (r = 0.45; p = 0.028) for the end-point "First day". The ROC analysis showed that mtDNA circulating in plasma (AUC = 0.605) can be a predictor of MOF development. The level of mtDNA significantly increases in case of MOF, irrespective of its cause. (2) The expression of mtDNA genes correlates with the level of MOF severity on the SOFA score.


Assuntos
Procedimentos Cirúrgicos Cardíacos , Insuficiência de Múltiplos Órgãos , Humanos , Insuficiência de Múltiplos Órgãos/etiologia , Insuficiência de Múltiplos Órgãos/genética , DNA Mitocondrial/genética , Procedimentos Cirúrgicos Cardíacos/efeitos adversos , Mitocôndrias , Ponte Cardiopulmonar/efeitos adversos
4.
Int J Mol Sci ; 23(16)2022 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-36012630

RESUMO

Sepsis is a critical condition characterized by increased levels of pro-inflammatory cytokines and proliferating cells such as neutrophils and macrophages in response to microbial pathogens. Such processes lead to an abnormal inflammatory response and multi-organ failure. MicroRNAs (miRNA) are single-stranded non-coding RNAs with the function of gene regulation. This means that miRNAs are involved in multiple intracellular pathways and thus contribute to or inhibit inflammation. As a result, their variable expression in different tissues and organs may play a key role in regulating the pathophysiological events of sepsis. Thanks to this property, miRNAs may serve as potential diagnostic and prognostic biomarkers in such life-threatening events. In this narrative review, we collect the results of recent studies on the expression of miRNAs in heart, blood, lung, liver, brain, and kidney during sepsis and the molecular processes in which they are involved. In reviewing the literature, we find at least 122 miRNAs and signaling pathways involved in sepsis-related organ dysfunction. This may help clinicians to detect, prevent, and treat sepsis-related organ failures early, although further studies are needed to deepen the knowledge of their potential contribution.


Assuntos
MicroRNAs , Sepse , Regulação da Expressão Gênica , Humanos , Macrófagos/metabolismo , MicroRNAs/metabolismo , Insuficiência de Múltiplos Órgãos/genética , Insuficiência de Múltiplos Órgãos/metabolismo , Sepse/complicações , Sepse/genética , Sepse/metabolismo
5.
J Clin Lab Anal ; 36(8): e24569, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35754113

RESUMO

BACKGROUND: Long noncoding RNA (lncRNA) cancer susceptibility candidate gene 2 (CASC2) inhibits inflammation and multi-organ dysfunction in various ways. The present study was intended to explore the potency of blood lncRNA CASC2 as a biomarker for sepsis management. METHODS: Totally, 184 sepsis patients and 30 healthy controls were enrolled. The reverse transcription-quantitative polymerase chain reaction was used to detect lncRNA CASC2 expression in peripheral blood mononuclear cell samples from the subjects. Mortality during 28 days was recorded in sepsis patients. RESULTS: LncRNA CASC2 was decreased in sepsis patients [median (interquartile range [IQR]): 0.473 (0.241-0.773)] by comparison to healthy controls [median (IQR): 1.019 (0.676-1.685)] (p < 0.001). In sepsis patients, lncRNA CASC2 was negatively correlated with Acute Physiology and Chronic Health Evaluation II (APACHE II) (p = 0.001), Sequential Organ Failure Assessment (SOFA) (p < 0.001), SOFA-respiratory system (p = 0.010), SOFA-coagulation (p = 0.020), SOFA-liver (p = 0.019), and SOFA-renal (p = 0.010) scores, but was not related to SOFA-nervous (p = 0.466) and SOFA-cardio vascular system (p = 0.059) scores. Additionally, lncRNA CASC2 was negatively related to tumor necrosis factor-α (p = 0.024), interleukin (IL)-1ß (p = 0.013), and IL-17A (p = 0.002), but was not linked to IL-6 (p = 0.112) or IL-10 (p = 0.074). Furthermore, lncRNA CASC2 was lower in sepsis deaths [median (IQR): 0.286 (0.166-0.475)] than in survivors [median (IQR): 0.534 (0.296-0.811)] (p < 0.001). Simultaneously, Kaplan-Meier (KM) curve analysis also observed that lncRNA CASC2 was inversely related to accumulating mortality in sepsis patients (p = 0.003). While lncRNA CASC2 could independently predict lower mortality risk. CONCLUSION: Circulating lncRNA CASC2 inadequacy indicates the release of inflammatory cytokines, severe multi-organ injuries, and increased mortality in sepsis patients.


Assuntos
RNA Longo não Codificante , Sepse , Biomarcadores , Citocinas , Humanos , Leucócitos Mononucleares , Insuficiência de Múltiplos Órgãos/genética , Prognóstico , RNA Longo não Codificante/genética , Índice de Gravidade de Doença , Proteínas Supressoras de Tumor
6.
J Clin Lab Anal ; 36(3): e24268, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35119126

RESUMO

OBJECTIVE: Long noncoding RNA plasmacytoma variant translocation 1 (lnc-PVT1) promotes septic inflammation and organ injuries via multiple ways, while its clinical engagement in sepsis management is indistinct. This study aimed to investigate its relationship with inflammation, multiple organ dysfunction, and mortality risk in sepsis patients. METHODS: Sepsis patients and age-/gender-matched healthy controls were enrolled; their lnc-PVT1 expression in plasma were detected by RT-qPCR. For sepsis patients only, the inflammatory cytokine levels (tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, and IL-17A) in plasma were detected by ELISA. According to the survival data during 28-day follow-up, sepsis patients were divided into sepsis survivors and sepsis deaths. RESULTS: Lnc-PVT1 expression was increased in sepsis patients (N = 157) compared with healthy controls (N = 80) (p < 0.001). In sepsis patients, lnc-PVT1 was linked with higher acute physiology and chronic health evaluation II (APACHEII) score (p = 0.001), total sequential organ failure assessment (SOFA) score, and its most subitems (SOFA-respiratory system, SOFA-coagulation, SOFA-liver, SOFA-cardiovascular system, and SOFA-renal system scores) (all p < 0.01), but not SOFA-nervous system score (p = 0.091); it did not relate to primary infection sites either (p = 0.204). Furthermore, lnc-PVT1 correlated with increased C-reactive protein, TNF-α, IL-1ß, and IL-17 in sepsis patients (all p < 0.01). Additionally, lnc-PVT1 expression was higher in sepsis deaths than that in sepsis survivors (p < 0.001), following receiver-operating characteristic curve disclosed that lnc-PVT1 predicted 28-day septic mortality risk (area under the curve: 0.789, 95% confidence interval: 0.702-0.875). CONCLUSION: Circulating lnc-PVT1 exhibits the potential as a biomarker in sepsis patients to inform inflammation, multiple organ dysfunction, and mortality risk.


Assuntos
RNA Longo não Codificante , Sepse , Biomarcadores , Humanos , Inflamação/genética , Insuficiência de Múltiplos Órgãos/genética , Prognóstico , RNA Longo não Codificante/genética , Curva ROC
7.
Ann Surg ; 275(6): e781-e788, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33427755

RESUMO

OBJECTIVE: The aim of this study was to identify genetic variants associated with early multiple organ failure (MOF) in acute pancreatitis. SUMMARY BACKGROUND DATA: MOF is a life-threatening complication of acute pancreatitis, and risk factors are largely unknown, especially in early persistent MOF. Genetic risk factors are thought to enhance severity in complex diseases such as acute pancreatitis. METHODS: A 2-phase study design was conducted. First, we exome sequenced 9 acute pancreatitis patients with early persistent MOF and 9 case-matched patients with mild edematous pancreatitis (phenotypic extremes) from our initial Dutch cohort of 387 patients. Secondly, 48 candidate variants that were overrepresented in MOF patients and 10 additional variants known from literature were genotyped in a replication cohort of 286 Dutch and German patients. RESULTS: Exome sequencing resulted in 161,696 genetic variants, of which the 38,333 non-synonymous variants were selected for downstream analyses. Of these, 153 variants were overrepresented in patients with multiple-organ failure, as compared with patients with mild acute pancreatitis. In total, 58 candidate variants were genotyped in the joined Dutch and German replication cohort. We found the rs12440118 variant of ZNF106 to be overrepresented in patients with MOF (minor allele frequency 20.4% vs 11.6%, Padj=0.026). Additionally, SLC52A1 rs346821 was found to be overrepresented (minor allele frequency 48.0% vs 42.4%, Padj= 0.003) in early MOF. None of the variants known from literature were associated.Conclusions: This study indicates that SLC52A1, a riboflavin plasma membrane transporter, and ZNF106, a zinc finger protein, may be involved in disease progression toward (early) MOF in acute pancreatitis.


Assuntos
Proteínas de Ligação a DNA , Pancreatite , Receptores Acoplados a Proteínas G , Humanos , Doença Aguda , Proteínas de Ligação a DNA/genética , Sequenciamento do Exoma , Insuficiência de Múltiplos Órgãos/genética , Pancreatite/complicações , Pancreatite/genética , Receptores Acoplados a Proteínas G/genética , Fatores de Risco , Dedos de Zinco
8.
Eur J Trauma Emerg Surg ; 48(3): 1545-1553, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32524156

RESUMO

PURPOSE: Overactive neutrophils are thought to be key drivers in the development of post-traumatic multiple organ dysfunction syndrome (MODS). Little is known about the role of inflammation-related lnc-IL7R in trauma. Thus, we aimed to explore the association between neutrophil-derived lnc-IL7R and post-traumatic MODS. METHODS: Total RNA was extracted from the isolated circulating neutrophils in 60 patients with trauma and 33 healthy volunteers for lnc-IL7R expression determination by real-time PCR. The correlation of lnc-IL7R expression with disease severity and the development of post-traumatic MODS was analyzed. RESULTS: The lnc-IL7R levels were significantly lower in trauma patients, especially in those with severe trauma [Injury Severity Score (ISS) ≥ 16], and correlated negatively with the ISS, Acute Physiology and Chronic Health Evaluation II score, and length of ICU stay. The lnc-IL7R levels were also significantly decreased in patients who developed MODS than in those who did not. Lnc-IL7R was an independent predictor of MODS [odds ratio (OR) 0.654, (0.435-0.982), p = 0.041]. The area under the curve for predicting post-traumatic MODS was 0.799 (sensitivity 76.9%, specificity 71.4%), with a cutoff value of 0.024. CONCLUSIONS: Neutrophil-derived lnc-IL7R is an independent predictor of post-traumatic MODS; therefore, it could be a useful predictive marker for MODS.


Assuntos
Traumatismo Múltiplo , RNA Longo não Codificante , Biomarcadores , Humanos , Escala de Gravidade do Ferimento , Insuficiência de Múltiplos Órgãos/genética , Traumatismo Múltiplo/complicações , Neutrófilos , RNA Longo não Codificante/genética
9.
Crit Care ; 25(1): 36, 2021 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-33494815

RESUMO

BACKGROUND: Sepsis is a life-threatening condition accompanied by organ dysfunction subsequent to a dysregulated host response to infection. Up to 60% of patients with sepsis develop acute kidney injury (AKI), which is associated with a poor clinical outcome. The pathophysiology of sepsis-associated AKI (sepsis-AKI) remains incompletely understood, but mitochondria have emerged as key players in the pathogenesis. Therefore, our aim was to identify mitochondrial damage in patients with sepsis-AKI. METHODS: We conducted a clinical laboratory study using "warm" postmortem biopsies from sepsis-associated AKI patients from a university teaching hospital. Biopsies were taken from adult patients (n = 14) who died of sepsis with AKI at the intensive care unit (ICU) and control patients (n = 12) undergoing tumor nephrectomy. To define the mechanisms of the mitochondrial contribution to the pathogenesis of sepsis-AKI, we explored mRNA and DNA expression of mitochondrial quality mechanism pathways, DNA oxidation and mitochondrial DNA (mtDNA) integrity in renal biopsies from sepsis-AKI patients and control subjects. Next, we induced human umbilical vein endothelial cells (HUVECs) with lipopolysaccharide (LPS) for 48 h to mimic sepsis and validate our results in vitro. RESULTS: Compared to control subjects, sepsis-AKI patients had upregulated mRNA expression of oxidative damage markers, excess mitochondrial DNA damage and lower mitochondrial mass. Sepsis-AKI patients had lower mRNA expression of mitochondrial quality markers TFAM, PINK1 and PARKIN, but not of MFN2 and DRP1. Oxidative DNA damage was present in the cytosol of tubular epithelial cells in the kidney of sepsis-AKI patients, whereas it was almost absent in biopsies from control subjects. Oxidative DNA damage co-localized with both the nuclei and mitochondria. Accordingly, HUVECs induced with LPS for 48 h showed an increased mnSOD expression, a decreased TFAM expression and higher mtDNA damage levels. CONCLUSION: Sepsis-AKI induces mitochondrial DNA damage in the human kidney, without upregulation of mitochondrial quality control mechanisms, which likely resulted in a reduction in mitochondrial mass.


Assuntos
Injúria Renal Aguda/genética , DNA Mitocondrial/análise , Rim/fisiopatologia , Sepse/genética , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/fisiopatologia , Adulto , Idoso , Dano ao DNA/fisiologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Insuficiência de Múltiplos Órgãos/etiologia , Insuficiência de Múltiplos Órgãos/genética , Sepse/complicações
10.
Clin Exp Immunol ; 203(3): 433-447, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33232517

RESUMO

Sepsis is an intractable clinical syndrome characterized by organ dysfunction when the body over-responds to an infection. Sepsis has a high fatality rate and lacks effective treatment. Family with sequence similarity 96 member A (FAM96A) is an evolutionarily conserved protein with high expression in the immune system and is related to cytosolic iron assembly and tumour suppression; however, research has been rarely conducted on its immune functions. Our study found that Fam96a-/- mice significantly resisted lesions during sepsis simulated by caecal ligation and puncture (CLP) or endotoxicosis models. After a challenge with lipopolysaccharide (LPS) or infection, Fam96a-/- mice exhibited less organ damage, longer survival and better bacterial clearance with decreased levels of proinflammatory cytokines. While screening several subsets of immune cells, FAM96A-expressing macrophages as the key cell type inhibited sepsis development. In-vivo macrophage depletion or adoptive transfer experiments abrogated significant differences in the survival of sepsis between Fam96a-/- and wild-type mice. Results of the bone marrow-derived macrophage (BMDM) polarization experiment indicated that FAM96A deficiency promotes the transformation of uncommitted monocytes/macrophages (M0) into M2 macrophages, secreting fewer proinflammatory cytokines. FAM96A may mediate an immunometabolism shift - from oxidative phosphorylation (OXPHOS) to glycolysis - in macrophages during sepsis, mirrored by reactive oxygen species (ROS) and glucose uptake. These data demonstrate that FAM96A regulates inflammatory response and provide a novel genomic insight for sepsis treatment.


Assuntos
Proteínas de Transporte/genética , Ativação de Macrófagos/genética , Macrófagos/metabolismo , Sepse/genética , Animais , Proteínas de Transporte/metabolismo , Linhagem Celular , Citocinas/metabolismo , Endotoxemia/induzido quimicamente , Endotoxemia/genética , Endotoxemia/metabolismo , Mediadores da Inflamação/metabolismo , Lipopolissacarídeos , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Insuficiência de Múltiplos Órgãos/genética , Insuficiência de Múltiplos Órgãos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Sepse/metabolismo , Análise de Sobrevida
11.
Crit Care Med ; 48(5): 745-756, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32167492

RESUMO

OBJECTIVES: Recent evidence from the fields of microbiology and immunology, as well as a small number of human sepsis studies, suggest that epigenetic regulation may play a central role in the pathogenesis of sepsis. The term "epigenetics" refers to regulatory mechanisms that control gene expression but are not related to changes in DNA sequence. These include DNA methylation, histone modifications, and regulation of transcription via non-coding RNAs. Epigenetic modifications, occurring in response to external stressors, lead to changes in gene expression, and thus lie at the intersection between genetics and the environment. In this review, we examine data from in vitro studies, animal studies, and the existing human sepsis studies in epigenetics to demonstrate that epigenetic mechanisms are likely central to the pathogenesis of sepsis and that epigenetic therapies may have potential in the treatment of sepsis and its associated organ failures. DATA SOURCES: Online search of published scientific literature via Pubmed using the term "epigenetics" in combination with the terms "sepsis", "infection", "bacterial infection", "viral infection", "critical illness", "acute respiratory distress syndrome", and "acute lung injury". STUDY SELECTION: Articles were chosen for inclusion based on their relevance to sepsis, acute inflammation, sepsis-related immune suppression, and sepsis-related organ failure. Reference lists were reviewed to identify additional relevant articles. DATA EXTRACTION: Relevant data was extracted and synthesized for narrative review. DATA SYNTHESIS: Epigenetic regulation is a key determinant of gene expression in sepsis. At the onset of infection, host-pathogen interactions often result in epigenetic alterations to host cells that favor pathogen survival. In parallel, the host inflammatory response is characterized by epigenetic modifications in key regulatory genes, including tumor necrosis factor and interleukin-1ß. In human sepsis patients, multiple epigenetic modifying enzymes show differential expression in early sepsis, suggesting a role for epigenetics in coordinating the response to infection. In the later stages of sepsis, epigenetic modifications accompany endotoxin tolerance and the immune-suppressed state. In animal models, treatment with epigenetic modifiers can mitigate the effects of sepsis and improve survival as well as reverse sepsis-associated organ injury. CONCLUSIONS: Epigenetic modifications are associated with key phases of sepsis, from the host-pathogen interaction, to acute inflammation, to immune suppression. Epigenetic markers show promise in the diagnosis and prognosis of sepsis and epigenetic modifying agents show promise as therapeutic tools in animal models of sepsis. Human studies in the area of epigenetics are sorely lacking and should be a priority for sepsis researchers.


Assuntos
Estado Terminal , Epigênese Genética/fisiologia , Sepse/genética , Sepse/fisiopatologia , Lesão Pulmonar Aguda/genética , Lesão Pulmonar Aguda/fisiopatologia , Animais , Biomarcadores , Metilação de DNA/fisiologia , Modelos Animais de Doenças , Expressão Gênica/fisiologia , Histonas/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Insuficiência de Múltiplos Órgãos/genética , Insuficiência de Múltiplos Órgãos/fisiopatologia , RNA não Traduzido/metabolismo , Síndrome do Desconforto Respiratório/genética , Síndrome do Desconforto Respiratório/fisiopatologia
13.
PLoS One ; 15(2): e0228503, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32074120

RESUMO

The plasma protein fetuin-A mediates the formation of protein-mineral colloids known as calciprotein particles (CPP)-rapid clearance of these CPP by the reticuloendothelial system prevents errant mineral precipitation and therefore pathological mineralization (calcification). The mutant mouse strain D2,Ahsg-/- combines fetuin-A deficiency with the calcification-prone DBA/2 genetic background, having a particularly severe compound phenotype of microvascular and soft tissue calcification. Here we studied mechanisms leading to soft tissue calcification, organ damage and death in these mice. We analyzed mice longitudinally by echocardiography, X-ray-computed tomography, analytical electron microscopy, histology, mass spectrometry proteomics, and genome-wide microarray-based expression analyses of D2 wildtype and Ahsg-/- mice. Fetuin-A-deficient mice had calcified lesions in myocardium, lung, brown adipose tissue, reproductive organs, spleen, pancreas, kidney and the skin, associated with reduced growth, cardiac output and premature death. Importantly, early-stage calcified lesions presented in the lumen of the microvasculature suggesting precipitation of mineral containing complexes from the fluid phase of blood. Genome-wide expression analysis of calcified lesions and surrounding (not calcified) tissue, together with morphological observations, indicated that the calcification was not associated with osteochondrogenic cell differentiation, but rather with thrombosis and fibrosis. Collectively, these results demonstrate that soft tissue calcification can start by intravascular mineral deposition causing microvasculopathy, which impacts on growth, organ function and survival. Our study underscores the importance of fetuin-A and related systemic regulators of calcified matrix metabolism to prevent cardiovascular disease, especially in dysregulated mineral homeostasis.


Assuntos
Calcinose/complicações , Calcinose/genética , Microvasos/patologia , Insuficiência de Múltiplos Órgãos/genética , Calcificação Vascular/genética , alfa-2-Glicoproteína-HS/genética , Animais , Calcinose/metabolismo , Calcinose/patologia , Cálcio/metabolismo , Doenças Cardiovasculares/genética , Doenças Cardiovasculares/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Knockout , Microcirculação/fisiologia , Microvasos/metabolismo , Minerais/metabolismo , Sistema Fagocitário Mononuclear/metabolismo , Sistema Fagocitário Mononuclear/patologia , Insuficiência de Múltiplos Órgãos/patologia , Calcificação Vascular/metabolismo , Calcificação Vascular/patologia , alfa-2-Glicoproteína-HS/deficiência
14.
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
15.
J Clin Invest ; 128(11): 5056-5072, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30320600

RESUMO

Dysregulated intestinal epithelial apoptosis initiates gut injury, alters the intestinal barrier, and can facilitate bacterial translocation leading to a systemic inflammatory response syndrome (SIRS) and/or multi-organ dysfunction syndrome (MODS). A variety of gastrointestinal disorders, including inflammatory bowel disease, have been linked to intestinal apoptosis. Similarly, intestinal hyperpermeability and gut failure occur in critically ill patients, putting the gut at the center of SIRS pathology. Regulation of apoptosis and immune-modulatory functions have been ascribed to Thirty-eight-negative kinase 1 (TNK1), whose activity is regulated merely by expression. We investigated the effect of TNK1 on intestinal integrity and its role in MODS. TNK1 expression induced crypt-specific apoptosis, leading to bacterial translocation, subsequent septic shock, and early death. Mechanistically, TNK1 expression in vivo resulted in STAT3 phosphorylation, nuclear translocation of p65, and release of IL-6 and TNF-α. A TNF-α neutralizing antibody partially blocked development of intestinal damage. Conversely, gut-specific deletion of TNK1 protected the intestinal mucosa from experimental colitis and prevented cytokine release in the gut. Finally, TNK1 was found to be deregulated in the gut in murine and porcine trauma models and human inflammatory bowel disease. Thus, TNK1 might be a target during MODS to prevent damage in several organs, notably the gut.


Assuntos
Proteínas Fetais/metabolismo , Doenças Inflamatórias Intestinais/enzimologia , Intestinos/enzimologia , Insuficiência de Múltiplos Órgãos/enzimologia , Traumatismo Múltiplo/enzimologia , Proteínas Tirosina Quinases/metabolismo , Síndrome de Resposta Inflamatória Sistêmica/enzimologia , Animais , Modelos Animais de Doenças , Feminino , Proteínas Fetais/genética , Doenças Inflamatórias Intestinais/etiologia , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/patologia , Interleucina-6/genética , Interleucina-6/metabolismo , Intestinos/patologia , Camundongos , Insuficiência de Múltiplos Órgãos/etiologia , Insuficiência de Múltiplos Órgãos/genética , Insuficiência de Múltiplos Órgãos/patologia , Traumatismo Múltiplo/complicações , Traumatismo Múltiplo/genética , Traumatismo Múltiplo/patologia , Proteínas Tirosina Quinases/genética , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Suínos , Síndrome de Resposta Inflamatória Sistêmica/etiologia , Síndrome de Resposta Inflamatória Sistêmica/patologia , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
16.
Clin Exp Immunol ; 194(1): 93-102, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29920655

RESUMO

Suppressor of cytokine signalling (SOCS) proteins are crucial negative regulators in many signalling pathways and are implicated in the pathogenesis of infectious diseases. The purpose of this study was to uncover possible associations of common polymorphisms within SOCS genes with infectious outcomes after traumatic injury. A total of 1087 trauma patients (Chongqing cohort 806 and Yunnan cohort 281) were recruited and followed-up for the development of infectious outcomes, such as sepsis and multiple organ dysfunction syndrome (MODS). Twelve selected single nucleotide polymorphisms (SNPs) were screened by pyrosequencing to determine their genotypes and associations with infectious complications. Among the 12 selected SNPs, only the cytokine-inducible Src homology (SH2) domain protein (CISH) promoter rs414171 polymorphism was found consistently to be associated statistically with the incidence of sepsis and MOD score in the two cohorts, despite analysing the SNPs independently or in combination. Further, patients with a T allele had significantly lower CISH expression and lower production of tumour necrosis factor (TNF)-α, but higher production of interleukin (IL)-10. Luciferase assay confirmed that the A→T variant in the rs414171 polymorphism inhibited the transcriptional activities of the CISH gene significantly. The CISH rs414171 polymorphism is associated significantly with susceptibility to sepsis and MODS in traumatic patients, which might prove to be a novel biomarker for indicating risk of infectious outcomes in critically injured patients.


Assuntos
Insuficiência de Múltiplos Órgãos/genética , Sepse/genética , Proteínas Supressoras da Sinalização de Citocina/genética , Ferimentos e Lesões/patologia , Adolescente , Adulto , Idoso , Feminino , Predisposição Genética para Doença/genética , Humanos , Interleucina-10/biossíntese , Masculino , Pessoa de Meia-Idade , Insuficiência de Múltiplos Órgãos/microbiologia , Polimorfismo de Nucleotídeo Único/genética , Regiões Promotoras Genéticas/genética , Estudos Prospectivos , Sepse/microbiologia , Fator de Necrose Tumoral alfa/biossíntese , Ferimentos e Lesões/microbiologia , Adulto Jovem
17.
Arterioscler Thromb Vasc Biol ; 38(8): 1772-1784, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29930006

RESUMO

Objective- Nbeal2-/- mice, a model of human gray platelet syndrome, have reduced neutrophil granularity and impaired host defense against systemic Staphylococcus aureus infection. We here aimed to study the role of Nbeal2 deficiency in both leukocytes and platelets during gram-negative pneumonia and sepsis. Approach and Results- We studied the role of Nbeal2 in platelets and leukocytes during murine pneumonia and sepsis by Klebsiella pneumoniae. Apart from platelet α-granule deficiency and reduced neutrophil granularity, also monocyte granularity was reduced in Nbeal2-/- mice, whereas plasma levels of MPO (myeloperoxidase), elastase, NGAL (neutrophil gelatinase-associated lipocalin), and MMP-9 (matrix metalloproteinase 9), and leukocyte CD11b expression were increased. Nbeal2-/- leukocytes showed unaltered in vitro antibacterial response and phagocytosis capacity against Klebsiella, and unchanged reactive nitrogen species and cytokine production. Also during Klebsiella pneumonia and sepsis, Nbeal2-/- mice had similar bacterial growth in lung and distant body sites, with enhanced leukocyte migration to the bronchoalveolar space. Despite similar infection-induced inflammation, organ damage was increased in Nbeal2-/- mice, which was also seen during endotoxemia. Platelet-specific Nbeal2 deficiency did not influence leukocyte functions, indicating that Nbeal2 directly modifies leukocytes. Transfusion of Nbeal2-/- but not of Nbeal2+/+ platelets into thrombocytopenic mice was associated with bleeding in the lung but similar host defense, pointing at a role for platelet α-granules in maintaining vascular integrity but not host defense during Klebsiella pneumosepsis. Conclusions- These data show that Nbeal2 deficiency-resulting in gray platelet syndrome-affects platelets, neutrophils, and monocytes, with intact host defense but increased organ damage during gram-negative pneumosepsis.


Assuntos
Plaquetas/metabolismo , Proteínas Sanguíneas/deficiência , Síndrome da Plaqueta Cinza/metabolismo , Infecções por Klebsiella/metabolismo , Klebsiella pneumoniae/patogenicidade , Insuficiência de Múltiplos Órgãos/metabolismo , Pneumonia Bacteriana/metabolismo , Sepse/metabolismo , Animais , Plaquetas/microbiologia , Proteínas Sanguíneas/genética , Antígeno CD11b/sangue , Modelos Animais de Doenças , Feminino , Síndrome da Plaqueta Cinza/sangue , Síndrome da Plaqueta Cinza/genética , Interações Hospedeiro-Patógeno , Infecções por Klebsiella/sangue , Infecções por Klebsiella/genética , Infecções por Klebsiella/microbiologia , Klebsiella pneumoniae/crescimento & desenvolvimento , Lipocalina-2/sangue , Masculino , Metaloproteinase 9 da Matriz/sangue , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/metabolismo , Monócitos/microbiologia , Insuficiência de Múltiplos Órgãos/sangue , Insuficiência de Múltiplos Órgãos/genética , Insuficiência de Múltiplos Órgãos/microbiologia , Neutrófilos/metabolismo , Neutrófilos/microbiologia , Elastase Pancreática/sangue , Peroxidase/sangue , Complexo Glicoproteico GPIb-IX de Plaquetas/genética , Complexo Glicoproteico GPIb-IX de Plaquetas/metabolismo , Transfusão de Plaquetas , Pneumonia Bacteriana/sangue , Pneumonia Bacteriana/genética , Pneumonia Bacteriana/microbiologia , Sepse/sangue , Sepse/genética , Sepse/microbiologia
18.
J Trauma Acute Care Surg ; 85(3): 580-587, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29847538

RESUMO

BACKGROUND: To investigate the effect of biliary tract external drainage (BTED) on inflammatory mediators and pathomorphism of intestine, liver, and lung in septic rats. METHOD: 48 SD rats (n = 8 per group) were randomized into six groups: control, sepsis, sepsis plus BTED, normal bile (obtained from eight healthy rats), and septic bile infusion for 6 hours respectively to test the effects of BTED bile infusion on cytokines' expression and tissue injury in the intestine, liver, and lung of septic/normal rats. Co-cultivation of intestinal epithelial cells (IEC-6) with bile for 12 hours was performed to evaluate the potential cytotoxicity of septic bile. Survival rate for sepsis plus BTED rats was detected compared with sepsis without BTED group (n = 20 per group) at 24, 48, and 72 hours, respectively. RESULTS: BTED for 6 hours significantly reduced the mRNA expression levels of tumor necrosis factor alpha (TNF-α) and IL-1ß (all p < 0.05 vs. sepsis group), whereas mRNA expression of TNF-α and IL-1ß in the intestine was increased after 6 hours' septic bile infusion compared with normal bile infusion group (all p < 0.05). TNF-α concentration in septic bile was significantly higher than that in the control group (p < 0.001). Tissue injury was significantly attenuated after 6 hours' BTED. CONCLUSIONS: BTED can significantly restrain the mRNA expression of TNF-α and IL-1ß in the intestine, liver, and lung and attenuate histological damage in septic rats.


Assuntos
Bile/metabolismo , Procedimentos Cirúrgicos do Sistema Biliar/métodos , Drenagem/métodos , Mediadores da Inflamação/metabolismo , Fígado/metabolismo , Pulmão/metabolismo , Sepse/metabolismo , Animais , Bile/fisiologia , Citocinas/metabolismo , Modelos Animais de Doenças , Interleucina-1beta/genética , Intestinos/fisiopatologia , Intestinos/cirurgia , Fígado/fisiopatologia , Pulmão/fisiopatologia , Masculino , Insuficiência de Múltiplos Órgãos/genética , Insuficiência de Múltiplos Órgãos/patologia , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley/genética , Sepse/patologia , Sepse/cirurgia , Fator de Necrose Tumoral alfa/genética
19.
Anesth Analg ; 126(1): 308-321, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28759485

RESUMO

Perioperative organ injury has a significant impact on surgical outcomes and presents a leading cause of death in the United States. Recent research has pointed out an important role of hypoxia signaling in the protection from organ injury, including for example myocardial infarction, acute respiratory distress syndrome, acute kidney, or gut injury. Hypoxia induces the stabilization of hypoxia-inducible factors (HIFs), thereby leading to the induction of HIF target genes, which facilitates adaptive responses to low oxygen. In this review, we focus on current therapeutic strategies targeting hypoxia signaling in various organ injury models and emphasize potential clinical approaches to integrate these findings into the care of surgical patients. Conceptually, there are 2 options to target the HIF pathway for organ protection. First, drugs became recently available that promote the stabilization of HIFs, most prominently via inhibition of prolyl hydroxylase. These compounds are currently trialed in patients, for example, for anemia treatment or prevention of ischemia and reperfusion injury. Second, HIF target genes (such as adenosine receptors) could be activated directly. We hope that some of these approaches may lead to novel pharmacologic strategies to prevent or treat organ injury in surgical patients.


Assuntos
Marcação de Genes/métodos , Hipóxia/metabolismo , Complicações Intraoperatórias/metabolismo , Insuficiência de Múltiplos Órgãos/metabolismo , Transdução de Sinais/fisiologia , Animais , Humanos , Fator 1 Induzível por Hipóxia/genética , Fator 1 Induzível por Hipóxia/metabolismo , Complicações Intraoperatórias/prevenção & controle , Insuficiência de Múltiplos Órgãos/genética , Insuficiência de Múltiplos Órgãos/prevenção & controle
20.
J Trauma Acute Care Surg ; 81(3): 478-85, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27306446

RESUMO

INTRODUCTION: The effect of sex on posttraumatic pathophysiology and outcomes after severe traumatic injury remains debated. We sought to determine the relationship of sex to the genomic and inflammatory responses, and clinical outcomes after hemorrhagic shock. METHODS: We analyzed blunt trauma patients in hemorrhagic shock from a prospective multi-institutional cohort study to assess for sex-based differences in the genomic response and clinical outcomes. Serially drawn blood samples were analyzed to evaluate peripheral leukocyte genomewide expression and circulating inflammatory mediators at intervals between 0.5 and 28 days after injury. Multivariate logistic regression models were developed to assess the effect of sex on outcomes after controlling for age, injury and shock severity, blood transfusion, and comorbidities. RESULTS: The cohort consisted of 1,285 (67%) male and 643 (33%) female blunt trauma patients. Injury and shock severity were similar between the two groups. There were small but statistically significant differences between males and females regarding their age, body mass index, and 12-hour blood and crystalloid administration. Organ failure was more severe in males, with slower recovery (9.0 vs. 6.5 days) in males compared to females (p < 0.01). However, there were no differences between males and females in plasma levels of IL-6, IL-8, IL-10, IL-1ß, tumor necrosis factor alpha, and monocyte chemoattractant protein 1. Multivariate analysis revealed that sex was not a significant independent risk factor for complicated recovery or 28-day mortality. Transcriptomic analysis revealed 333 genes with significant differential expression patterns between males and females (FDR, <0.001), including genes associated with general inflammation, innate immunity, cell adhesion, and cell signaling. None of the former genes were directly associated with sex hormones or X/Y chromosomes. CONCLUSION: There are sex-specific differences in the leukocyte genomic response to severe injury that are associated with more robust and longer-duration organ dysfunction. However, these expression patterns do not seem to be associated with sex-linked genes or circulating cytokine level differences, and do not translate to worsened sex-specific organ failure outcomes or inpatient mortality. LEVEL OF EVIDENCE: Prognostic/epidemiologic study, level III.


Assuntos
Genômica , Imunidade Inata , Insuficiência de Múltiplos Órgãos/genética , Insuficiência de Múltiplos Órgãos/imunologia , Choque Hemorrágico/genética , Choque Hemorrágico/imunologia , Ferimentos não Penetrantes/genética , Ferimentos não Penetrantes/imunologia , Adulto , Citocinas/sangue , Feminino , Humanos , Escala de Gravidade do Ferimento , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Fatores de Risco , Fatores Sexuais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA