ABSTRACT
Ocular involvement in systemic diseases is frequent in cats; however, without concurrent clinical and ophthalmic examinations with gross and/or histologic analysis of the eye, these findings can be underdiagnosed. This article aims to provide gross, histologic, and immunohistochemical characteristics of ocular lesions from cats submitted to necropsy, focusing on those caused by systemic infectious agents. Cats that died due to a systemic infectious disease were selected based on necropsy diagnosis and presence of ocular lesions. Gross, histologic, and immunohistochemical findings were recorded. From April 2018 to September 2019, 849 eyes of 428 cats were evaluated. Histologic abnormalities were seen in 29% of cases, which were classified as inflammatory (41%), neoplastic (32%), degenerative (19%), and metabolic/vascular (8%). Macroscopic changes were present in one-third of eyes with histologic lesions. Of these, 40% were attributed to inflammatory or neoplastic diseases associated with infectious agents. The most important infectious agents causing ocular disease in this study were feline leukemia virus, feline infectious peritonitis virus, and Cryptococcus sp. The most common ocular abnormalities associated with infectious agents were uveitis (anterior, posterior, or panuveitis), optic neuritis, and meningitis of the optic nerve. Ocular lesions secondary to systemic infections in cats are frequent; however, these are not always diagnosed because gross lesions are less common than histologic lesions. Therefore, both gross and histologic evaluation of the eyes of cats is recommended, mainly for cases in which the clinical suspicion or necropsy diagnosis suggests that an infectious agent might be related to the cause of death.
Subject(s)
Cat Diseases , Communicable Diseases , Feline Infectious Peritonitis , Neoplasms , Sepsis , Uveitis , Cats , Animals , Eye/pathology , Uveitis/pathology , Uveitis/veterinary , Neoplasms/pathology , Neoplasms/veterinary , Sepsis/pathology , Sepsis/veterinary , Communicable Diseases/pathology , Communicable Diseases/veterinary , Cat Diseases/pathology , Feline Infectious Peritonitis/pathologyABSTRACT
Abstract Sepsis is described as a life-threatening organ dysfunction caused by a host's response to infection, leading to an unbalance in body homeostasis. It is one of the leading causes of death in developed countries. Considering that in critically ill patients, such as those with sepsis, plasma concentrations do not necessarily reflect tissue concentrations, one way to assess tissue concentrations is through the microdialysis technique, which allows direct measurements of free drug at the site of action. This review was carried out after searching the Pubmed, Scielo and Web of Science databases, using the following descriptors: (microdialysis AND (sepsis OR septic shock OR severe sepsis OR septicemia)) OR (microdialysis AND (sepsis OR septic shock OR severe sepsis) OR septicemia) AND (antimicrobial OR antibiotic OR antifungal)). The physiological changes generated by sepsis may imply changes in pharmacokinetic parameters, such as in clearance, which may be reduced in these patients and in volume of distribution, which presents an expansion, mainly due to edema. Both events contribute to a high inter- individual variability in tissue penetration of antimicrobials which is generally observed in patients with sepsis.
Subject(s)
Pharmaceutical Preparations/supply & distribution , Microdialysis/methods , Sepsis/pathology , Pharmacokinetics , Pharmacologic Actions , Anti-Infective Agents/pharmacologyABSTRACT
Abstract Neonatal sepsis continues to be a major cause of morbidity and mortality worldwide. Coagulase-negative staphylococci (CoNS), commonly found on the skin, being the main agents isolated. The aim of this study was to evaluate CoNS isolated from blood cultures of newborn (NB) infants. The study took place between 2014 and 2016/2017 in a tertiary hospital in southern Brazil. Using the VITEK 2 system (bioMérieux, Marcy l'Etoile, France), the microorganisms were identified and had their sensitivity profiles determined. The minimum inhibitory concentrations of linezolid, tigecycline, and vancomycin were also determined. The clinical parameters and mortality rates of NBs were evaluated. From January to December 2014, 176 CoNS isolates were obtained from 131 patients and from June 2016 to July 2017, 120 CoNS isolates were obtained from 79 patients. Staphylococcus epidermidis was most prevalent in both periods. Resistance rates increased between 2014 and 2016/2017, especially against ciprofloxacin (52.27% and 73.11%, p = 0.0004), erythromycin (51.40% and 68.07%, p = 0.0054), gentamicin (50.59% and 67.23%, p = 0.0052), and penicillin (71.3% and 99.17%, p = 0.0001), respectively. With 100% susceptibility to linezolid, tigecycline, and vancomycin in both periods and methodologies tested. In 2014, 53.44% of the NBs received antibiotic therapy, and of these, 77.14% used a catheter; in 2016/2017, these were 78.48% and 95.16%, respectively. Regarding laboratory tests, a hemogram was ineffective, since patients with sepsis presented normal reference values. In 2014 and 2016/17, 15.71% and 17.74% of the NBs died, respectively. S. epidermidis was the predominant microorganism, related to catheter use in most cases. The resistance rates have increased over time, demonstrating the importance of adopting control and prevention measures in this hospital. CoNS are responsible for a significant neonatal sepsis mortality rate in infants.
Subject(s)
Humans , Male , Female , Infant, Newborn , Staphylococcal Scalded Skin Syndrome/pathology , Infant, Newborn , Coagulase/adverse effects , Skin , Staphylococcus epidermidis/pathogenicity , Microbial Sensitivity Tests/instrumentation , Mortality , Sepsis/pathology , Blood Culture/classification , Blood Culture/instrumentation , HospitalsABSTRACT
Abstract RGX-365 is the main fraction of black ginseng conmprising protopanaxatriol (PPT)-type rare ginsenosides (ginsenosides Rg4, Rg6, Rh4, Rh1, and Rg2). No studies on the antiseptic activity of RGX-365 have been reported. High mobility group box 1 (HMGB1) is recognized as a late mediator of sepsis, and the inhibition of HMGB1 release and recovery of vascular barrier integrity have emerged as attractive therapeutic strategies for the management of sepsis. In this study, we examined the effects of RGX-365 on HMGB1-mediated septic responses and survival rate in a mouse sepsis model. RGX-365 was administered to the mice after HMGB1 challenge. The antiseptic activity of RGX-365 was assessed based on the production of HMGB1, measurement of permeability, and septic mouse mortality using a cecal ligation and puncture (CLP)-induced sepsis mouse model and HMGB1-activated human umbilical vein endothelial cells (HUVECs). We found that RGX-365 significantly reduced HMGB1 release from LPS- activated HUVECs and CLP-induced release of HMGB1 in mice. RGX-365 also restored HMGB1-mediated vascular disruption and inhibited hyperpermeability in the mice. In addition, treatment with RGX-365 reduced sepsis-related mortality in vivo. Our results suggest that RGX- 365 reduces HMGB1 release and septic mortality in vivo, indicating that it is useful in the treatment of sepsis.
Subject(s)
HMGB1 Protein/analysis , Panax/adverse effects , Permeability , Sepsis/pathology , Ginsenosides , Human Umbilical Vein Endothelial Cells/classification , Anti-Infective Agents, Local/adverse effectsSubject(s)
Ecthyma/immunology , Primary Immunodeficiency Diseases/diagnosis , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/isolation & purification , Sepsis/immunology , Skin/pathology , Ecthyma/diagnosis , Ecthyma/pathology , Fatal Outcome , Gangrene , Humans , Infant , Interleukin-1 Receptor-Associated Kinases , Male , Primary Immunodeficiency Diseases/complications , Pseudomonas Infections/diagnosis , Pseudomonas Infections/pathology , Sepsis/diagnosis , Sepsis/pathology , Skin/immunology , Skin/microbiologyABSTRACT
Sepsis and septic shock are associated with acute and sustained impairment in the function of the cardiovascular system, kidneys, lungs, liver, and brain, among others. Despite the significant advances in prevention and treatment, sepsis and septic shock sepsis remain global health problems with elevated mortality rates. Rho proteins can interact with a considerable number of targets, directly affecting cellular contractility, actin filament assembly and growing, cell motility and migration, cytoskeleton rearrangement, and actin polymerization, physiological functions that are intensively impaired during inflammatory conditions, such as the one that occurs in sepsis. In the last few decades, Rho proteins and their downstream pathways have been investigated in sepsis-associated experimental models. The most frequently used experimental design included the exposure to bacterial lipopolysaccharide (LPS), in both in vitro and in vivo approaches, but experiments using the cecal ligation and puncture (CLP) model of sepsis have also been performed. The findings described in this review indicate that Rho proteins, mainly RhoA and Rac1, are associated with the development of crucial sepsis-associated dysfunction in different systems and cells, including the endothelium, vessels, and heart. Notably, the data found in the literature suggest that either the inhibition or activation of Rho proteins and associated pathways might be desirable in sepsis and septic shock, accordingly with the cellular system evaluated. This review included the main findings, relevance, and limitations of the current knowledge connecting Rho proteins and sepsis-associated experimental models.
Subject(s)
Sepsis/enzymology , Shock, Septic/enzymology , rho GTP-Binding Proteins/metabolism , Animals , Disease Models, Animal , Humans , Molecular Targeted Therapy , Sepsis/drug therapy , Sepsis/pathology , Shock, Septic/drug therapy , Shock, Septic/pathology , Signal Transduction , rac1 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/agonists , rho GTP-Binding Proteins/antagonists & inhibitors , rhoA GTP-Binding Protein/metabolismABSTRACT
Acute lung injury and acute respiratory distress syndrome can occur as a result of sepsis. Cardiac dysfunction is a serious component of multi-organ failure caused by severe sepsis. Telomere shortening is related to several heart diseases. Telomeres are associated with the shelterin protein complex, which contributes to the maintenance of telomere length. Low-power infrared lasers modulate mRNA levels of shelterin complex genes. This study aimed to evaluate effects of a low-power infrared laser on mRNA relative levels of genes involved in telomere stabilization and telomere length in heart tissue of an experimental model of acute lung injury caused by sepsis. Animals were divided into six groups, treated with intraperitoneal saline solution, saline solution and exposed to a low-power infrared laser at 10 J cm-2 and 20 J cm-2, lipopolysaccharide (LPS), and LPS and, after 4 h, exposed to a low-power infrared laser at 10 J cm-2 and 20 J cm-2. The laser exposure was performed only once. Analysis of mRNA relative levels and telomere length by RT-qPCR was performed. Telomere shortening and reduction in mRNA relative levels of TRF1 mRNA in heart tissues of LPS-induced ALI animals were observed. In addition, laser exposure increased the telomere length at 10 J cm-2 and modulated the TRF1 mRNA relative levels of at 20 J cm-2 in healthy animals. Although the telomeres were shortened and mRNA levels of TRF1 gene were increased in nontreated controls, the low-power infrared laser irradiation increased the telomere length at 10 J cm-2 in cardiac tissue of animals affected by LPS-induced acute lung injury, which suggests that telomere maintenance is a part of the photobiomodulation effect induced by infrared radiation.
Subject(s)
Acute Lung Injury/genetics , Heart , Lasers , Sepsis/genetics , Telomere/genetics , Acute Lung Injury/chemically induced , Acute Lung Injury/pathology , Animals , Lipopolysaccharides , RNA, Messenger/genetics , Sepsis/chemically induced , Sepsis/pathologyABSTRACT
Sepsis-induced myocardial dysfunction considerably increases mortality risk in patients with sepsis. Previous studies from our group have shown that sepsis alters the expression of structural proteins in cardiac cells, resulting in cardiomyocyte degeneration and impaired communication between cardiac cells. Caveolin-3 (CAV3) is a structural protein present in caveolae, located in the membrane of cardiac muscle cells, which regulates physiological processes such as calcium homeostasis. In sepsis, there is a disruption of calcium homeostasis, which increases the concentration of intracellular calcium, which can lead to the activation of potent cellular enzymes/proteases which cause severe cellular injury and death. The purpose of the present study was to test the hypotheses that sepsis induces CAV3 overexpression in the heart, and the regulation of L-type calcium channels directly relates to the regulation of CAV3 expression. Severe sepsis increases the expression of CAV3 in the heart, as immunostaining in our study showed CAV3 presence in the cardiomyocyte membrane and cytoplasm, in comparison with our control groups (without sepsis) that showed CAV3 presence predominantly in the plasma membrane. The administration of verapamil, an L-type calcium channel inhibitor, resulted in a decrease in mortality rates of septic mice. This effect was accompanied by a reduction in the expression of CAV3 and attenuation of cardiac lesions in septic mice treated with verapamil. Our results indicate that CAV3 has a vital role in cardiac dysfunction development in sepsis and that the regulation of L-type calcium channels may be related to its expression.
Subject(s)
Caveolin 3/metabolism , Heart/drug effects , Sepsis/drug therapy , Verapamil/therapeutic use , Animals , Calcium Channels, L-Type , Humans , Male , Mice , Sepsis/mortality , Sepsis/pathology , Survival Analysis , Verapamil/pharmacologyABSTRACT
BACKGROUND: Sepsis is an emergency medical condition that can lead to death and it is defined as a life-threatening organ dysfunction caused by immune dysregulation in response to an infection. It is considered the main killer in intensive care units. Sepsis associated-encephalopathy (SAE) is mostly caused by a sepsis-induced systemic inflammatory response. Studies report SAE in 14-63% of septic patients. Main SAE symptoms are not specific and usually include acute impairment of consciousness, delirium and/or coma, along with electroencephalogram (EEG) changes. For those who recover from sepsis and SAE, impaired cognitive function, mobility and quality of life are often observed months to years after hospital discharge, and there is no treatment available today to prevent that. Inflammation and oxidative stress are key players for the SAE pathophysiology. Gold nanoparticles have been demonstrated to own important anti-inflammatory properties. It was also reported 20 nm citrate-covered gold nanoparticles (cit-AuNP) reduce oxidative stress. In this context, we tested whether 20 nm cit-AuNP could alleviate the acute changes caused by sepsis in brain of mice, with focus on inflammation. Sepsis was induced in female C57BL/6 mice by cecal ligation and puncture (CLP), 20 nm cit-AuNP or saline were intravenously (IV) injected 2 h after induction of sepsis and experiments performed 6 h after induction. Intravital microscopy was used for leukocyte and platelet adhesion study in brain, blood brain barrier (BBB) permeability carried out by Evans blue assay, cytokines measured by ELISA and real time PCR, cell adhesion molecules (CAMs) by flow cytometry and immunohistochemistry, and transcription factors, by western blotting. RESULTS: 20 nm cit-AuNP treatment reduced leukocyte and platelet adhesion to cerebral blood vessels, prevented BBB failure, reduced TNF- concentration in brain, and ICAM-1 expression both in circulating polymorphonuclear (PMN) leukocytes and cerebral blood vessels of mice with sepsis. Furthermore, 20 nm cit-AuNP did not interfere with the antibiotic effect on the survival rate of mice with sepsis. CONCLUSIONS: Cit-AuNP showed important anti-inflammatory properties in the brain of mice with sepsis, being a potential candidate to be used as adjuvant drug along with antibiotics in the treatment of sepsis to avoid SAE.
Subject(s)
Cecum/metabolism , Gold/pharmacology , Inflammation/drug therapy , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Microvessels/metabolism , Sepsis/drug therapy , Animals , Blood-Brain Barrier/metabolism , Brain/metabolism , Brain/pathology , Brain Diseases , Cell Adhesion , Cytokines/metabolism , Disease Models, Animal , Female , Inflammation/pathology , Leukocytes/metabolism , Mice , Mice, Inbred C57BL , Microvessels/pathology , Neutrophils/metabolism , Quality of Life , Sepsis/metabolism , Sepsis/pathology , Sepsis-Associated Encephalopathy/metabolismABSTRACT
Sepsis is characterized by a dysregulated immune response to infection characterized by an early hyperinflammatory and oxidative response followed by a subsequent immunosuppression phase. Although there have been some advances in the treatment of sepsis, mortality rates remain high, urging for the search of new therapies. ß-Lapachone (ß-Lap) is a natural compound obtained from Tabebuia avellanedae Lorentz ex Griseb. with several pharmacological properties including bactericidal, anti-inflammatory, and antioxidant activity. Thus, the aim of this study was to evaluate the effects of ß-Lap in a mouse sepsis model. To this, we tested two therapeutic protocols in mice submitted to cecal ligation and puncture- (CLP-) induced sepsis. First, we found that in pretreated animals, ß-Lap reduced the systemic inflammatory response and improved bacterial clearance and mouse survival. Moreover, ß-Lap also decreased lipid peroxidation and increased the total antioxidant capacity in the serum and peritoneal cavity of septic animals. In the model of severe sepsis, the posttreatment with ß-Lap was able to increase the survival of animals and maintain the antioxidant defense function. In conclusion, the ß-Lap was able to increase the survival of septic animals by a mechanism involving immunomodulatory and antioxidant protective effects.
Subject(s)
Naphthoquinones/therapeutic use , Sepsis/drug therapy , Sepsis/mortality , Animals , Anti-Inflammatory Agents/therapeutic use , Chemoprevention/methods , Cytokines/metabolism , Disease Models, Animal , Immunosuppression Therapy/methods , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/mortality , Inflammation Mediators/metabolism , Male , Mice , Oxidative Stress/drug effects , Sepsis/metabolism , Sepsis/pathology , Survival RateABSTRACT
Sepsis is a severe disease characterized by an uncontrolled systemic inflammation and consequent organ dysfunction generated in response to an infection. Extracellular ATP acting through the P2X7 receptor induces the maturation and release of pro-inflammatory cytokines (i.e., IL-1ß) and the production of reactive nitrogen and oxygen species that lead to oxidative tissue damage. Here, we investigated the role of the P2X7 receptor in inflammation, oxidative stress, and liver injury in sepsis. Sepsis was induced by cecal ligation and puncture (CLP) in wild-type (WT) and P2X7 knockout (P2X7-/-) mice. The oxidative stress in the liver of septic mice was assessed by 2',7'-dichlorofluorescein oxidation reaction (DCF), thiobarbituric acid-reactive substances (TBARS), and nitrite levels dosage. The status of the endogenous defense system was evaluated through catalase (CAT) and superoxide dismutase (SOD) activities. The inflammation was assessed histologically and by determining the expression of inflammatory cytokines and chemokines by RT-qPCR. We observed an increase in the reactive species and lipid peroxidation in the liver of septic WT mice, but not in the liver from P2X7-/- animals. We found an imbalance SOD/CAT ratio, also only WT septic animals. The number of inflammatory cells and the gene expression of IL-1 ß, IL-6, TNF-α, IL-10, CXCL1, and CXCL2 were higher in the liver of WT septic mice in comparison to P2X7-/- septic animals. In summary, our results suggest that the P2X7 receptor might be a therapeutic target to limit oxidative stress damage and liver injury during sepsis.
Subject(s)
Liver Diseases/metabolism , Oxidative Stress/physiology , Receptors, Purinergic P2X7/metabolism , Sepsis/metabolism , Sepsis/pathology , Animals , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, KnockoutSubject(s)
COVID-19/pathology , Embolism/pathology , Endocarditis, Bacterial/pathology , Endocarditis/pathology , Respiratory Distress Syndrome/pathology , Sepsis/pathology , Adult , Angiography , COVID-19/complications , COVID-19/diagnostic imaging , COVID-19/virology , Embolism/complications , Embolism/diagnostic imaging , Embolism/virology , Endocarditis/complications , Endocarditis/diagnostic imaging , Endocarditis/virology , Endocarditis, Bacterial/complications , Endocarditis, Bacterial/diagnostic imaging , Endocarditis, Bacterial/virology , Fatal Outcome , Humans , Lung/diagnostic imaging , Lung/pathology , Lung/virology , Male , Respiration, Artificial , Respiratory Distress Syndrome/complications , Respiratory Distress Syndrome/diagnostic imaging , Respiratory Distress Syndrome/virology , SARS-CoV-2/growth & development , SARS-CoV-2/pathogenicity , Sepsis/complications , Sepsis/diagnostic imaging , Sepsis/virology , Tomography, X-Ray ComputedABSTRACT
Only in the last decade the long-term consequences of sepsis started to be studied and even less attention has been given to the treatment of psychological symptoms of sepsis survivors. It is estimated that 60% of sepsis survivors have psychological disturbances, including depression, anxiety, and cognitive impairment. Although the causative factors remain largely poorly understood, blood-brain barrier (BBB) disturbances, neuroinflammation, and oxidative stress have been investigated. Therefore, we sought to explore if the immunomodulatory and antioxidant selenocompound 3-[(4-chlorophenyl)selanyl]-1-methyl-1H-indole (CMI) would be able to ameliorate long-term behavioral and biochemical alterations in sepsis survivors male Swiss mice. CMI treatment (1 mg/kg, given orally for seven consecutive days) attenuated depression- and anxiogenic-like behaviors and cognitive impairment present one month after the induction of sepsis (lipopolysaccharide, 5 mg/kg intraperitoneally). Meantime, CMI treatment modulated the number of neutrophils and levels of reactive species in neutrophils, lymphocytes, and monocytes. In addition, peripheral markers of liver and kidneys dysfunction (AST, ALT, urea, and creatinine) were reduced after CMI treatment in post-septic mice. Notably, CMI treatment to non-septic mice did not alter AST, ALT, urea, and creatinine levels, indicating the absence of acute hepatotoxicity and nephrotoxicity following CMI treatment. Noteworthy, CMI ameliorated BBB dysfunction induced by sepsis, modulating the expression of inflammation-associated genes (NFκB, IL-1ß, TNF-α, IDO, COX-2, iNOS, and BDNF) and markers of oxidative stress (reactive species, nitric oxide, and lipid peroxidation levels) in the prefrontal cortices and hippocampi of mice. In conclusion, we unraveled crucial molecular pathways that are impaired in post-septic mice and we present CMI as a promising therapeutic candidate aimed to manage the long-lasting behavioral alterations of sepsis survivors to improve their quality of life.
Subject(s)
Behavior, Animal , Indoles/chemistry , Oxidative Stress , Sepsis/pathology , Animals , Anxiety/drug therapy , Anxiety/etiology , Behavior, Animal/drug effects , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Depression/drug therapy , Depression/etiology , Depression/pathology , Disease Models, Animal , Indoles/pharmacology , Indoles/therapeutic use , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Kidney/drug effects , Kidney/metabolism , Lipopolysaccharides/toxicity , Liver/drug effects , Liver/metabolism , Locomotion/drug effects , Male , Mice , Neutrophils/cytology , Neutrophils/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Sepsis/complicationsABSTRACT
Sepsis-associated encephalopathy causes brain dysfunction that can result in cognitive impairments in sepsis survivor patients. In previous work, we showed that simvastatin attenuated oxidative stress in brain structures related to memory in septic rats. However, there is still a need to evaluate the long-term impact of simvastatin administration on brain neurodegenerative processes and cognitive damage in sepsis survivors. Here, we investigated the possible neuroprotective role of simvastatin in neuroinflammation, and neurodegeneration conditions of brain structures related to memory in rats at 10 days after sepsis survival. Male Wistar rats (250-300 g) were submitted to cecal ligation and puncture (CLP, n = 42) or remained as non-manipulated (naïve, n = 30). Both groups were treated (before and after the surgery) by gavage with simvastatin (20 mg/kg) or an equivalent volume of saline and observed for 10 days. Simvastatin-treated rats that survived to sepsis showed a reduction in the levels of nitrate, IL1-ß, and IL-6 and an increase in Bcl-2 protein expression in the prefrontal cortex and hippocampus, and synaptophysin only in the hippocampus. Immunofluorescence revealed a reduction of glial activation, neurodegeneration, apoptosis, and amyloid aggregates confirmed by quantification of GFAP, Iba-1, phospho Ser396-tau, total tau, cleaved caspase-3, and thioflavin-S in the prefrontal cortex and hippocampus. In addition, treated animals presented better performance in tasks involving habituation memory, discriminative, and aversive memory. These results suggest that statins exert a neuroprotective role by upregulation of the Bcl-2 and gliosis reduction, which may prevent the cognitive deficit observed in sepsis survivor animals.
Subject(s)
Brain/drug effects , Cognitive Dysfunction/prevention & control , Neurodegenerative Diseases/drug therapy , Sepsis/drug therapy , Simvastatin/therapeutic use , Animals , Brain/metabolism , Brain/pathology , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Male , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Rats , Rats, Wistar , Sepsis/metabolism , Sepsis/pathology , Simvastatin/pharmacologyABSTRACT
PURPOSE: The objective of this study was to investigate the accuracy of 18F-FDG-PET in the diagnosis of multibacterial abdominal sepsis by cecum ligation and puncture (CLP) in rats. METHODS: Adult Wistar rats ( Rattus norvegicus ), weighing 227±35g, were allocated into a sepsis group by CLP (n=10) and sham group (n=10). 18F-FDG-PET using microPET was performed on all rats after 24 hours. RESULTS: All animals survived for postoperative 24h. The abdomen/liver ratio of the standardized uptake value (SUV) percentage was significantly higher in the sepsis group than in the sham (p=0.004). The ROC curve showed an accuracy of 18F-FDG-PET to detect abdominal sepsis of 88.9% (p=0.001), sensitivity of 90% and specificity of 88.9%. When a cut-off point of 79% of the ratio between the SUV on the abdominal region and liver was established, the sensitivity was 90%, specificity of 88.9%; positive and negative predictive values of 90.0% and 88.9%, respectively. CONCLUSIONS: The diagnostic accuracy of 18F-FDG-PET in rats with abdominal sepsis was significantly high. It was also demonstrated the predictive ability of the abdomen/liver SUV ratio to diagnose abdominal sepsis. These findings may have implications for the clinical setting, locating septic foci with PETscan.
Subject(s)
Fluorodeoxyglucose F18 , Intraabdominal Infections/diagnostic imaging , Positron-Emission Tomography/methods , Radiopharmaceuticals , Sepsis/diagnostic imaging , Animals , Intraabdominal Infections/pathology , Predictive Value of Tests , Rats , Rats, Wistar , Reference Values , Reproducibility of Results , Sepsis/pathology , Time FactorsABSTRACT
BACKGROUND: Previous data have reported that the growth of established tumors may be facilitated by postsepsis disorder through changes in the microenvironment and immune dysfunction. However, the influence of postsepsis disorder in initial carcinogenesis remains elusive. METHODS: In the present work, the effect of postsepsis on inflammation-induced early carcinogenesis was evaluated in an experimental model of colitis-associated colorectal cancer (CAC). We also analyzed the frequency and role of intestinal T regulatory cells (Treg) in CAC carcinogenesis. RESULTS: The colitis grade and the tumor development rate were evaluated postmortem or in vivo through serial colonoscopies. Sepsis-surviving mice (SSM) presented with a lower colonic DNA damage, polyp incidence, reduced tumor load, and milder colitis than their sham-operated counterparts. Ablating Treg led to restoration of the ability to develop colitis and tumor polyps in the SSM, in a similar fashion to that in the sham-operated mice. On the other hand, the growth of subcutaneously inoculated MC38luc colorectal cancer cells or previously established chemical CAC tumors was increased in SSM. CONCLUSION: Our results provide evidence that postsepsis disorder has a dual effect in cancer development, inhibiting inflammation-induced early carcinogenesis in a Treg-dependent manner, while increasing the growth of previously established tumors.
Subject(s)
Colitis/complications , Colonic Neoplasms/pathology , Disease Models, Animal , Inflammation/complications , Sepsis/complications , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment/immunology , Animals , Colitis/immunology , Colitis/pathology , Colonic Neoplasms/etiology , Cytokines/metabolism , Female , Inflammation/pathology , Mice , Mice, Inbred C57BL , Sepsis/immunology , Sepsis/pathology , Signal TransductionABSTRACT
INTRODUCTION: Various pathologies and lifestyle factors, such as nutritional factors and physical exercises, can alter the gene expression of proteins related to synthesis and degradation. AIM: We performed a systematic review of atrophy models, cancer models, burn models, sepsis models, cardiac insufficiency models, amino acid supplementation models, protein supplementation models, and miscellaneous models that have altered the gene expression of MTOR, MURF-1, or MAFBX in rats and mice. MATERIALS AND METHODS: We searched the literature in the following databases: Medline, Scielo.org, Scielo.br, Redib, Lilacs, and the Periodicos Capes. RESULTS: We selected 56 articles for this review. DISCUSSION: Several conditions can alter the gene expression of muscle proteins under conditions that stimulate muscle degradation pathways. Therefore, treatments must normalize the expression of the degradation pathways and potentiate the synthesis pathways so the muscular tissue confers an increase in functional capacity and thus, survival in diseased patients. Therefore, the reversal of the mechanisms that promote its depletion must be achieved. CONCLUSION: Identification of the atrophic mechanisms present in pathologies and other conditions of muscular disuse in the scientific literature is fundamental for the adoption of clinical strategies to prevent protein degradation and to promote the maintenance and/or increase of muscle tissue. Such strategies include physical exercise, protein supplementation, and/or pharmacological applications, aimed toward restoring the fullness of functional capacity.
Subject(s)
Gene Expression Regulation/genetics , Muscle Proteins/genetics , SKP Cullin F-Box Protein Ligases/genetics , TOR Serine-Threonine Kinases/genetics , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/pathology , Animals , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/pathology , Atrophy/genetics , Atrophy/pathology , Burns/genetics , Burns/pathology , Disease Models, Animal , Humans , Mice , Neoplasms/genetics , Neoplasms/pathology , Rats , Sepsis/genetics , Sepsis/pathologyABSTRACT
INTRODUCTION: Sepsis is an important cause of mortality and morbidity, and inflammatory response and oxidative stress play major roles underlying its pathophysiology. Here, we evaluated the effect of intraperitoneal etanercept administration on oxidative stress and inflammation indicators in the kidney and blood of experimental sepsis-induced rats. METHODS: Twenty-eight adult Sprague Dawley rats were classified into Control (Group 1), Sepsis (Group 2), Sepsis+Cefazolin (Group 3), and Sepsis+Cefazolin+Etanercept (Group 4) groups. Kidney tissue and serum samples were obtained for biochemical and histopathological investigations and examined for the C reactive protein (CRP), tumor necrosis factor-alpha (TNF-α), triggering receptor expressed on myeloid cells (TREM), and malondialdehyde (MDA) levels. RESULTS: The levels of TNF-α, TREM, and MDA in serum and kidney samples were significantly higher in rats from sepsis group than in rats from control group (p < 0.05). Group 3 showed a significant reduction in serum levels of TNF-α, CRP, and TREM as compared with Group 2 (p < 0.05). Serum TNF-α, CRP, TREM, and MDA levels and kidney TNF-α and TREM levels were significantly lower in Group 4 than in Group 2 (p < 0.05). Serum TNF-α and TREM levels in Group 4 were significantly lower than those in Group 3, and histopathological scores were significantly lower in Group 3 and Group 4 than in Group 2 (p < 0.05). Histopathological scores of Group 4 were significantly lower than those of Group 3 (p < 0.05). CONCLUSIONS: Etanercept, a TNF-α inhibitor, may ameliorate sepsis-induced oxidative stress, inflammation, and histopathological damage.
Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Etanercept/administration & dosage , Inflammation/prevention & control , Kidney/drug effects , Oxidative Stress/drug effects , Sepsis/pathology , Tumor Necrosis Factor-alpha/blood , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Disease Models, Animal , Etanercept/pharmacology , Inflammation/pathology , Injections, Intraperitoneal , Rats , Rats, Sprague-Dawley , Sepsis/bloodABSTRACT
BACKGROUND: In order to modulate microglial phenotypes in vivo, M1 microglia were depleted by administration of gadolinium chloride and the expression of M2 microglia was induced by IL-4 administration in an animal model of sepsis to better characterize the role of microglial phenotypes in sepsis-induced brain dysfunction. METHODS: Wistar rats were submitted to sham or cecal ligation and perforation (CLP) and treated with IL-4 or GdCl3. Animals were submitted to behavioral tests 10 days after surgery. In a separated cohort of animals at 24 h, 3 and 10 days after surgery, hippocampus was removed and cytokine levels, M1/M2 markers and CKIP-1 levels were determined. RESULTS: Modulation of microglia by IL-4 and GdCl3 was associated with an improvement in long-term cognitive impairment. When treated with IL-4 and GdCl3, the reduction of pro-inflammatory cytokines was apparent in almost all analyzed time points. Additionally, CD11b and iNOS were increased after CLP at all time points, and both IL-4 and GdCl3 treatments were able to reverse this. There was a significant decrease in CD11b gene expression in the CLP+GdCl3 group. IL-4 treatment was able to decrease iNOS expression after sepsis. Furthermore, there was an increase of CKIP-1 in the hippocampus of GdCl3 and IL-4 treated animals 10 days after CLP induction. CONCLUSIONS: GdCl3 and IL-4 are able to manipulate microglial phenotype in an animal models of sepsis, by increasing the polarization toward an M2 phenotype IL-4 and GdCl3 treatment was associated with decreased brain inflammation and functional recovery.
Subject(s)
Anti-Inflammatory Agents/pharmacology , Behavior, Animal/drug effects , Cognition/drug effects , Cognitive Dysfunction/prevention & control , Encephalitis/prevention & control , Gadolinium/pharmacology , Hippocampus/drug effects , Interleukin-4/pharmacology , Microglia/drug effects , Sepsis/drug therapy , Animals , CD11b Antigen/metabolism , Carrier Proteins/metabolism , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Cognitive Dysfunction/psychology , Cytokines/metabolism , Disease Models, Animal , Encephalitis/metabolism , Encephalitis/pathology , Encephalitis/physiopathology , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Inflammation Mediators/metabolism , Microglia/metabolism , Microglia/pathology , Nitric Oxide Synthase Type II/metabolism , Phenotype , Rats, Wistar , Sepsis/metabolism , Sepsis/pathology , Sepsis/physiopathology , Time FactorsABSTRACT
Escherichia coli is an important pathogen responsible for a variety of diseases. We have recently shown that Pic, a serine protease secreted by E. coli, mediates immune evasion by the direct cleavage of complement molecules. The aim of this study was to investigate the action of a Pic-producing bacteria in a murine model of sepsis. Mice were infected with Pic-producing E. coli (F5) or F5∆pic mutant. Animal survival was monitored for five days, and a subset of mice was euthanized after 12 h for sample acquisition. The inoculation of Pic-producing bacteria induced 100% death within 24 h. The colony forming units count in the organs was significantly higher in F5. Hematological analysis showed a decrease of total leukocytes. Nitric oxide and cytokines were detected in serum, as well as on peritoneal lavage of the F5 group in higher levels than those detected in the other groups. In addition, immunophenotyping showed a decrease of activated lymphocytes and macrophages in the F5 group. Therefore, Pic represents an important virulence factor, allowing the survival of the bacterium in the bloodstream and several organs, as well as inducing a high production of proinflammatory mediators by the host, and concomitantly a cellular immunosuppression, leading to sepsis and death.