RÉSUMÉ
In Chile, Piscirickettsia salmonis contains two genetically isolated genogroups, LF-89 and EM-90. However, the impact of a potential co-infection with these two variants on Salmonid Rickettsial Septicemia (SRS) in Atlantic salmon (Salmo salar) remains largely unexplored. In our study, we evaluated the effect of P. salmonis LF-89-like and EM-90-like co-infection on post-smolt Atlantic salmon after an intraperitoneal challenge to compare changes in disease dynamics and host immune response. Co-infected fish had a significantly lower survival rate (24.1%) at 21 days post-challenge (dpc), compared with EM-90-like single-infected fish (40.3%). In contrast, all the LF-89-like single-infected fish survived. In addition, co-infected fish presented a higher presence of clinical lesions than any of the single-infected fish. The gene expression of salmon immune-related biomarkers evaluated in the head kidney, spleen, and liver showed that the EM-90-like isolate and the co-infection induced the up-regulation of cytokines (e.g., il-1ß, ifnγ, il8, il10), antimicrobial peptides (hepdicin) and pattern recognition receptors (PRRs), such as TLR5s. Furthermore, in serum samples from EM-90-like and co-infected fish, an increase in the total IgM level was observed. Interestingly, specific IgM against P. salmonis showed greater detection of EM-90-like antigens in LF-89-like infected fish serum (cross-reaction). These data provide evidence that P. salmonis LF-89-like and EM-90-like interactions can modulate SRS disease dynamics in Atlantic salmon, causing a synergistic effect that increases the severity of the disease and the mortality rate of the fish. Overall, this study contributes to achieving a better understanding of P. salmonis population dynamics.
Sujet(s)
Co-infection , Maladies des poissons , Piscirickettsia , Infections à Piscirickettsiaceae , Salmo salar , Animaux , Piscirickettsia/physiologie , Maladies des poissons/microbiologie , Maladies des poissons/immunologie , Infections à Piscirickettsiaceae/médecine vétérinaire , Infections à Piscirickettsiaceae/microbiologie , Co-infection/médecine vétérinaire , Co-infection/microbiologie , Co-infection/immunologie , Chili , Sepsie/médecine vétérinaire , Sepsie/microbiologie , Sepsie/immunologieRÉSUMÉ
Sepsis is a complex condition of inflammatory and immune dysregulation, triggered by severe infection. In survivors, chronic inflammation and immune dysregulation linger, facilitating the emergence of infections. CD8 dysfunction contributes to immunosuppression in sepsis survivors. We devised an animal model that enabled us to identify and analyze CD8-intrinsic defects induced by sepsis. We adoptively transferred CD45.1 CD8 OT-I T cells into CD45.2 congenic mice and subjected them to cecal ligature and puncture, to induce abdominal sepsis. One month later, we isolated the transferred CD8 cells. Surface marker expression confirmed they had not been activated through the TCR. CD8 OT-I T cells isolated from septic (or sham-operated) mice were transferred to second recipients, which were challenged with OVA-expressing Listeria monocytogenes. We compared effector capacities between OT-I cells exposed to sepsis and control cells. Naive mice that received OT-I cells exposed to sepsis had higher bacterial burden and a shorter survival when challenged with OVA-expressing L. monocytogenes. OT-I cells isolated from septic mice produced less IFN-γ but had conserved activation, expansion potential, and cytotoxic function. We observed lower transcript levels of IFN-γ and of the long noncoding RNA Ifng-as1, a local regulator of the epigenetic landscape, in cells exposed to sepsis. Accordingly, local abundance of a histone modification characteristic of active promoter regions was reduced in sepsis-exposed CD8 T cells. Our results identify a mechanism through which inflammation in the context of sepsis affects CD8 T cell function intrinsically.
Sujet(s)
Lymphocytes T CD8+ , Chromatine , Interféron gamma , Listeria monocytogenes , Sepsie , Animaux , Souris , Transfert adoptif , Lymphocytes T CD8+/immunologie , Chromatine/immunologie , Chromatine/métabolisme , Modèles animaux de maladie humaine , Interféron gamma/immunologie , Listeria monocytogenes/immunologie , Infections à Listeria/immunologie , Activation des lymphocytes/immunologie , Souris de lignée C57BL , Sepsie/immunologieRÉSUMÉ
Sepsis treatment is a challenging condition due to its complexity, which involves host inflammatory responses to a severe and potentially fatal infection, associated with organ dysfunction. The aim of this study was to analyze the scientific literature on the immunomodulatory effects of glucans in a murine model of systemic infection induced by cecal ligation and puncture. This study comprises an integrative literature review based on systematic steps, with searches carried out in the PubMed, ScienceDirect, Scopus, Web of Science, and Embase databases. In most studies, the main type of glucan investigated was ß-glucan, at 50 mg/kg, and a reduction of inflammatory responses was identified, minimizing the occurrence of tissue damage leading to increased animal survival. Based on the data obtained and discussed in this review, glucans represent a promising biotechnological alternative to modulate the immune response and could potentially be used in the clinical management of septic individuals.
Sujet(s)
Modèles animaux de maladie humaine , Sepsie , Animaux , Sepsie/traitement médicamenteux , Sepsie/immunologie , Sepsie/thérapie , Humains , Souris , Glucanes/usage thérapeutique , Glucanes/pharmacologie , bêta-Glucanes/usage thérapeutique , Immunomodulation/effets des médicaments et des substances chimiquesRÉSUMÉ
Sepsis remains one of the main causes of death in intensive care unit (ICU) worldwide, despite all technological and scientific advances. Microvesicles (MV) have become promising biomarkers for quick and accurate monitoring of several illnesses. The aim of this pilot study was to characterize and evaluate the performance of MV as biomarker of clinical outcome in septic and trauma patients. For this purpose, 39 subjects, both genders, aging from 18 to 85 years were included in three groups referred as Sepsis, Trauma and Healthy Control. Kinetic analysis of MV was carried out at four consecutive time points: admission (baseline)/T1, 24 h/T2, 72 h/T3 and outcome/T4 of discharge or death. At admission, an overall increase in total MV (Annexin V+) was observed in Sepsis.MV CD14+ (monocytes) was a putative biomarker to identify trauma patients, while MV CD3+ (T-cells) and CD41+ (platelets) were qualified to discriminated Trauma from Sepsis. Sepsis (Death) presented an increase in MV Annexin V+, CD45+, CD16+, CD14+, and CD41+ in comparison to Sepsis (Discharge). Moreover, Trauma (Death) presented an increase of MV CD3+ and CD235+ as compared to Trauma (Discharge). Analysing the ROC curve of specific MV evaluated according to performance, an accuracy of 100% was found to segregate the outcome in sepsis, and 95% in trauma. Our findings suggest that MV might be useful as a potential role in discriminating outcome in patients with sepsis/septic shock and trauma with high accuracy. However, further studies with a larger number of participants will be necessary to validate our findings.
Sujet(s)
Marqueurs biologiques , Microparticules membranaires , Sepsie/sang , Plaies et blessures/sang , Adolescent , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Antigènes CD/immunologie , Maladie grave , Femelle , Humains , Mâle , Adulte d'âge moyen , Projets pilotes , Pronostic , Études rétrospectives , Sepsie/immunologie , Plaies et blessures/immunologie , Jeune adulteRÉSUMÉ
Sepsis is a global health emergency, which is caused by various sources of infection that lead to changes in gene expression, protein-coding, and metabolism. Advancements in "omics" technologies have provided valuable tools to unravel the mechanisms involved in the pathogenesis of this disease. In this study, we performed shotgun mass spectrometry in peripheral blood mononuclear cells (PBMC) from septic patients (N=24) and healthy controls (N=9) and combined these results with two public microarray leukocytes datasets. Through combination of transcriptome and proteome profiling, we identified 170 co-differentially expressed genes/proteins. Among these, 122 genes/proteins displayed the same expression trend. Ingenuity Pathway Analysis revealed pathways related to lymphocyte functions with decreased status, and defense processes that were predicted to be strongly increased. Protein-protein interaction network analyses revealed two densely connected regions, which mainly included down-regulated genes/proteins that were related to the transcription of RNA, translation of proteins, and mitochondrial translation. Additionally, we identified one module comprising of up-regulated genes/proteins, which were mainly related to low-density neutrophils (LDNs). LDNs were reported in sepsis and in COVID-19. Changes in gene expression level were validated using quantitative real-time PCR in PBMCs from patients with sepsis. To further support that the source of the upregulated module of genes/proteins found in our results were derived from LDNs, we identified an increase of this population by flow cytometry in PBMC samples obtained from the same cohort of septic patients included in the proteomic analysis. This study provides new insights into a reprioritization of biological functions in response to sepsis that involved a transcriptional and translational shutdown of genes/proteins, with exception of a set of genes/proteins related to LDNs and host-defense system.
Sujet(s)
Agranulocytes/métabolisme , Granulocytes neutrophiles/métabolisme , Sepsie/métabolisme , Bases de données factuelles , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes , Humains , Agranulocytes/cytologie , Cellules myéloïdes suppressives/cytologie , Cellules myéloïdes suppressives/métabolisme , Granulocytes neutrophiles/cytologie , Cartes d'interactions protéiques , Protéomique , Sepsie/génétique , Sepsie/immunologieSujet(s)
Ecthyma/immunologie , Maladies d'immunodéficience primaire/diagnostic , Infections à Pseudomonas/immunologie , Pseudomonas aeruginosa/isolement et purification , Sepsie/immunologie , Peau/anatomopathologie , Ecthyma/diagnostic , Ecthyma/anatomopathologie , Issue fatale , Gangrène , Humains , Nourrisson , Interleukin-1 Receptor-Associated Kinases , Mâle , Maladies d'immunodéficience primaire/complications , Infections à Pseudomonas/diagnostic , Infections à Pseudomonas/anatomopathologie , Sepsie/diagnostic , Sepsie/anatomopathologie , Peau/immunologie , Peau/microbiologieRÉSUMÉ
Sepsis is one of the most common comorbidities observed in diabetic patients, associated with a deficient innate immune response. Recently, we have shown that glucagon possesses anti-inflammatory properties. In this study, we investigated if hyperglucagonemia triggered by diabetes might reduce the migration of neutrophils, increasing sepsis susceptibility. 21 days after diabetes induction by intravenous injection of alloxan, we induced moderate sepsis in Swiss-Webster mice through cecum ligation and puncture (CLP). The glucagon receptor (GcgR) antagonist des-his1-[Glu9]-glucagon amide was injected intraperitoneally 24h and 1h before CLP. We also tested the effect of glucagon on CXCL1/KC-induced neutrophil migration to the peritoneal cavity in mice. Neutrophil chemotaxis in vitro was tested using transwell plates, and the expression of total PKA and phospho-PKA was evaluated by western blot. GcgR antagonist restored neutrophil migration, reduced CFU numbers in the peritoneal cavity and improved survival rate of diabetic mice after CLP procedure, however, the treatment did no alter hyperglycemia, CXCL1/KC plasma levels and blood neutrophilia. In addition, glucagon inhibited CXCL1/KC-induced neutrophil migration to the peritoneal cavity of non-diabetic mice. Glucagon also decreased the chemotaxis of neutrophils triggered by CXCL1/KC, PAF, or fMLP in vitro. The inhibitory action of glucagon occurred in parallel with the reduction of CXCL1/KC-induced actin polymerization in neutrophils in vitro, but not CD11a and CD11b translocation to cell surface. The suppressor effect of glucagon on CXCL1/KC-induced neutrophil chemotaxis in vitro was reversed by pre-treatment with GcgR antagonist and adenylyl cyclase or PKA inhibitors. Glucagon also increased PKA phosphorylation directly in neutrophils in vitro. Furthermore, glucagon impaired zymosan-A-induced ROS production by neutrophils in vitro. Human neutrophil chemotaxis and adherence to endothelial cells in vitro were inhibited by glucagon treatment. According to our results, this inhibition was independent of CD11a and CD11b translocation to neutrophil surface or neutrophil release of CXCL8/IL-8. Altogether, our results suggest that glucagon may be involved in the reduction of neutrophil migration and increased susceptibility to sepsis in diabetic mice. This work collaborates with better understanding of the increased susceptibility and worsening of sepsis in diabetics, which can contribute to the development of new effective therapeutic strategies for diabetic septic patients.
Sujet(s)
Mouvement cellulaire/effets des médicaments et des substances chimiques , Diabète expérimental/complications , Prédisposition aux maladies/étiologie , Glucagon/administration et posologie , Granulocytes neutrophiles/effets des médicaments et des substances chimiques , Sepsie/étiologie , Sepsie/immunologie , Adulte , Animaux , Mouvement cellulaire/immunologie , Chimiotaxie des leucocytes/effets des médicaments et des substances chimiques , Diabète expérimental/immunologie , Diabète expérimental/microbiologie , Femelle , Glucagon/métabolisme , Humains , Mâle , Souris , Lignées consanguines de souris , Granulocytes neutrophiles/immunologieRÉSUMÉ
Infectious diseases may affect brain function and cause encephalopathy even when the pathogen does not directly infect the central nervous system, known as infectious disease-associated encephalopathy. The systemic inflammatory process may result in neuroinflammation, with glial cell activation and increased levels of cytokines, reduced neurotrophic factors, blood-brain barrier dysfunction, neurotransmitter metabolism imbalances, and neurotoxicity, and behavioral and cognitive impairments often occur in the late course. Even though infectious disease-associated encephalopathies may cause devastating neurologic and cognitive deficits, the concept of infectious disease-associated encephalopathies is still under-investigated; knowledge of the underlying mechanisms, which may be distinct from those of encephalopathies of non-infectious cause, is still limited. In this review, we focus on the pathophysiology of encephalopathies associated with peripheral (sepsis, malaria, influenza, and COVID-19), emerging therapeutic strategies, and the role of neuroinflammation.
Sujet(s)
Encéphalopathies/immunologie , COVID-19/complications , Cytokines/immunologie , Grippe humaine/complications , Paludisme/complications , Sepsie/complications , Barrière hémato-encéphalique/immunologie , Encéphalopathies/prévention et contrôle , COVID-19/immunologie , Humains , Grippe humaine/immunologie , Paludisme/immunologie , Sepsie/immunologieRÉSUMÉ
BACKGROUND: The term sepsis is used to designate a systemic condition of infection and inflammation associated with hemodynamic changes that result in organic dysfunction. Gestational sepsis can impair the development of the central nervous system and may promote permanent behavior alterations in the offspring. The aim of our work was to evaluate the effects of maternal sepsis on inflammatory cytokine levels and synaptic proteins in the hippocampus, neocortex, frontal cortex, and cerebellum of neonatal, young, and adult mice. Additionally, we analyzed the motor development, behavioral features, and cognitive impairments in neonatal, young and adult offspring. METHODS: Pregnant mice at the 14th embryonic day (E14) were intratracheally instilled with saline 0.9% solution (control group) or Klebsiella spp. (3 × 108 CFU) (sepsis group) and started on meropenem after 5 h. The offspring was sacrificed at postnatal day (P) 2, P8, P30, and P60 and samples of liver, lung, and brain were collected for TNF-α, IL-1ß, and IL-6 measurements by ELISA. Synaptophysin, PSD95, and ß-tubulin levels were analyzed by Western blot. Motor tests were performed at all analyzed ages and behavioral assessments were performed in offspring at P30 and P60. RESULTS: Gestational sepsis induces a systemic pro-inflammatory response in neonates at P2 and P8 characterized by an increase in cytokine levels. Maternal sepsis induced systemic downregulation of pro-inflammatory cytokines, while in the hippocampus, neocortex, frontal cortex, and cerebellum an inflammatory response was detected. These changes in the brain immunity were accompanied by a reduction of synaptophysin and PSD95 levels in the hippocampus, neocortex, frontal cortex, and cerebellum, in all ages. Behavioral tests demonstrated motor impairment in neonates, and depressive-like behavior, fear-conditioned memory, and learning impairments in animals at P30 and P60, while spatial memory abilities were affected only at P60, indicating that gestational sepsis not only induces an inflammatory response in neonatal mouse brains, but also affects neurodevelopment, and leads to a plethora of behavioral alterations and cognitive impairments in the offspring. CONCLUSION: These data suggest that maternal sepsis may be causatively related to the development of depression, learning, and memory impairments in the litter.
Sujet(s)
Encéphale/immunologie , Effets différés de l'exposition prénatale à des facteurs de risque/immunologie , Sepsie/immunologie , Animaux , Comportement animal , Encéphale/métabolisme , Dysfonctionnement cognitif/étiologie , Femelle , Inflammation , Souris , Activité motrice/physiologie , Grossesse , Effets différés de l'exposition prénatale à des facteurs de risque/métabolisme , Sepsie/complications , Synapses/métabolismeRÉSUMÉ
Introduction: Sepsis has pro- and anti-inflammatory processes caused by infectious agents. Sepsis survivors have impaired immune response due to immunosuppression. Gene expression during the inflammatory process is guided by transcriptional access to chromatin, with post-translational changes made in histones that determine whether the loci of the inflammatory gene are active, balanced, or suppressed. For this, a review literature was performed in PubMed included 'sepsis' and 'epigenetic' and 'immunosuppression' terms until May 2020.Areas covered: This review article explores the relationship between epigenetic mechanisms and the pathophysiology of sepsis. Epigenetic changes, vulnerable gene expression, and immunosuppression are related to inflammatory insults that can modify the dynamics of the central nervous system. Therefore, it is important to investigate the timing of these changes and their dynamics during the disease progression.Expert opinion: Epigenetic changes are associated with the main stages of sepsis, from the pathogen-host interaction to inflammation and immunosuppression. These changes are key regulators of gene expression during physiological and pathological conditions. Thus, epigenetic markers have significant prognostic and diagnostic potential in sepsis, and epigenetic changes can be explored in combination with therapeutic strategies in experimental models of the disease.
Sujet(s)
Épigenèse génétique , Tolérance immunitaire , Sepsie , Épigenèse génétique/génétique , Épigenèse génétique/immunologie , Expression des gènes/génétique , Expression des gènes/immunologie , Histone/génétique , Histone/immunologie , Humains , Tolérance immunitaire/génétique , Tolérance immunitaire/immunologie , Mitochondries/immunologie , Mitochondries/métabolisme , Sepsie/génétique , Sepsie/immunologie , Sepsie/physiopathologieRÉSUMÉ
Natural killer (NK) cells have the dual ability to produce pro-inflammatory (IFNγ) and anti-inflammatory (IL-10) cytokines during systemic infection, which points to their crucial role both as inflammatory effectors for infection clearance and as regulators to counterbalance inflammation to limit immune-mediated damage to the host. In particular, immunosuppressive IL-10 secretion by NK cells has been described to occur in systemic, but not local, infections as a recent immunoregulatory mechanism of inflammation that may be detrimental or beneficial, depending on the timing of release, type of disease, or the infection model. Understanding the factors that drive the production of IL-10 by NK cells and their impact during dualistic inflammatory states, such as sepsis and other non-controlled inflammatory diseases, is relevant for achieving effective therapeutic advancements. In this review, the evidence regarding the immunoregulatory role of IL-10-producing NK cells in systemic infection is summarized and discussed in detail, and the potential molecular mechanisms that drive IL-10 production by NK cells are considered.
Sujet(s)
Interleukine-10/immunologie , Cellules tueuses naturelles/immunologie , Sepsie/immunologie , Animaux , Humains , Inflammation/immunologieRÉSUMÉ
Salmonid Rickettsial Septicaemia (SRS), caused by Piscirickettsia salmonis, is a severe bacterial disease in the Chilean salmon farming industry. Vaccines and antibiotics are the current strategies to fight SRS; however, the high frequency of new epizootic events confirms the need to develop new strategies to combat this disease. An innovative opportunity is perturbing the host pathways used by the microorganisms to replicate inside host cells through host-directed antimicrobial drugs (HDAD). Iron is a critical nutrient for P. salmonis infection; hence, the use of iron-chelators becomes an excellent alternative to be used as HDAD. The aim of this work was to use the iron chelator Deferiprone (DFP) as HDAD to treat SRS. Here, we describe the protective effect of the iron chelator DFP over P. salmonis infections at non-antibiotic concentrations, in bacterial challenges both in vitro and in vivo. At the cellular level, our results indicate that DFP reduced the intracellular iron content by 33.1% and P. salmonis relative load during bacterial infections by 78%. These findings were recapitulated in fish, where DFP reduced the mortality of rainbow trout challenged with P. salmonis in 34.9% compared to the non-treated group. This is the first report of the protective capacity of an iron chelator against infection in fish, becoming a potential effective host-directed therapy for SRS and other animals against ferrophilic pathogens.
Sujet(s)
Maladies des poissons/prévention et contrôle , Fer/pharmacologie , Oncorhynchus mykiss , Piscirickettsia/physiologie , Infections à Piscirickettsiaceae/médecine vétérinaire , Salmo salar , Sepsie/médecine vétérinaire , Phénomènes physiologiques nutritionnels chez l'animal , Animaux , Lignée cellulaire , Maladies des poissons/immunologie , Maladies des poissons/microbiologie , Fer/composition chimique , Infections à Piscirickettsiaceae/immunologie , Infections à Piscirickettsiaceae/microbiologie , Infections à Piscirickettsiaceae/prévention et contrôle , Sepsie/immunologie , Sepsie/microbiologie , Sepsie/prévention et contrôleRÉSUMÉ
Right now the world is going through an unprecedented pandemic caused by a novel coronavirus. Recent papers pointed out the fatal outcome in most of the severe cases, in which a cytokine storm has been proven to be the cause of a systemic shock, acute respiratory syndrome, multiorgan failure and consequently death. Several explanations have been proposed trying to explain the pathophysiology of the cytokine storm, but viral proteins with a possible superantigen activity as a cause of immune dysregulation have not been addressed. If this hypothesis is proven, a different treatment approach might change the outcome in severe cases.
Sujet(s)
Lymphocytes T CD4+/immunologie , COVID-19/immunologie , Cellules dendritiques/immunologie , SARS-CoV-2/physiologie , Sepsie/immunologie , Superantigènes/immunologie , Présentation d'antigène , Cytokines/métabolisme , Humains , PandémiesRÉSUMÉ
High levels of extracellular 72 kDa heat shock protein (eHSP72) can be detected in the serum of septic patients and are associated with increased oxidative profiles and elevated rates of mortality among these patients. However, a possible immunomodulatory role for this protein, resulting in tissue protection during sepsis, has never been assessed. In this study, we investigated whether eHSP72 administration could attenuate the severity of sepsis in a mouse peritonitis model. Animals (90-day-old male C57BL/6J mice) were divided into Sepsis (n = 8) and Sepsis + eHSP72 (n = 9) groups, which both received injections of 20% fecal solution [1 mg/g body weight (wt), intraperitoneal (i.p.)], to trigger peritonitis induced-sepsis, whereas a Control group (n = 7) received a saline injection. eHSP72 was administered (1.33 ng/g body wt) to the Sepsis+eHSP72 group, 12 h after sepsis induction. All animals were evaluated for murine sepsis score (MSS), hemogram, core temperature, and glycemia (before and 4, 12, and 24 h after sepsis induction). Treatment with eHSP72 promoted reduced sepsis severity 24 h after sepsis induction, based on MSS scores (Control = 1.14 ± 1.02; Sepsis = 11.07 ± 7.24, and Sepsis + eHSP72 = 5.62 ± 1.72, P < 0.001) and core temperatures (°C; Control = 37.48 ± 0.58; Sepsis = 35.17 ± 2.88, and Sepsis + eHSP72 = 36.94 ± 2.02; P = 0.006). eHSP72 treatment also limited the oxidative profile and respiratory dysfunction in mice with sepsis. Although sepsis modified glycemic levels and white and red blood cell counts, these variables were not influenced by eHSP72 treatment (P > 0.05). Finally, eHSP72 improved the survival rate after sepsis (P = 0.0371). Together, our results indicated that eHSP72 may ameliorate sepsis severity and possibly improve some sepsis indices in mice.
Sujet(s)
Protéines du choc thermique HSP72/administration et posologie , Péritonite/thérapie , Sepsie/thérapie , Animaux , Modèles animaux de maladie humaine , Immunomodulation , Perfusions veineuses , Mâle , Souris de lignée C57BL , Péritonite/immunologie , Sepsie/immunologieRÉSUMÉ
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.
Sujet(s)
Colite/complications , Tumeurs du côlon/anatomopathologie , Modèles animaux de maladie humaine , Inflammation/complications , Sepsie/complications , Lymphocytes T régulateurs/immunologie , Microenvironnement tumoral/immunologie , Animaux , Colite/immunologie , Colite/anatomopathologie , Tumeurs du côlon/étiologie , Cytokines/métabolisme , Femelle , Inflammation/anatomopathologie , Souris , Souris de lignée C57BL , Sepsie/immunologie , Sepsie/anatomopathologie , Transduction du signalRÉSUMÉ
Evidences has suggested that in the early life the innate immune system presents plasticity and the time and dose-adequate stimuli in this phase may program long-lasting immunological responses that persist until adulthood. We aimed to evaluate whether LPS challenge in early childhood period may modulate brain alterations after sepsis in adult life. Experiments were performed to evaluate the LPS challenge in early childhood or adult period on acute and long-term brain alterations after model of sepsis by cecal ligation and perforation (CLP) in adult life. Wistar rats were divided in saline+sham, LPS+sham, saline+CLP and LPS+CLP groups to determine cytokine levels and nitrite/nitrate concentration in cerebrospinal fluid (CSF); oxidative damage, activity of antioxidant enzymes (superoxide dismutase-SOD and catalase-CAT); blood brain barrier (BBB) permeability; myeloperoxidase (MPO) and epigenetic enzymes activities in the hippocampus and prefrontal cortex (at 24 h after CLP) and cognitive function, survival and brain-derived neurotrophic factor (BDNF) level (at ten days after CLP). LPS-preconditioning in early life could lead to decreased levels of TNF-α and IL-6 and oxidative damage parameters in the brain after CLP in adult rats. In addition, LPS-preconditioning in early life increase CAT activity, attenuates the BBB permeability and epigenetic enzymes alterations and in long term, improves the memory, BDNF levels and survival. In conclusion, rats submitted to CLP in adulthood displayed acute neuroinflammation, neurochemical and epigenetic alteration improvement accompanied in long term by an increase in survival, neurotrophin level and memory performance when preconditioned with LPS in the early life.
Sujet(s)
Encéphale/immunologie , Médiateurs de l'inflammation/métabolisme , Lipopolysaccharides/toxicité , Neuro-immunomodulation/immunologie , Neuroprotection/immunologie , Sepsie/immunologie , Facteurs âges , Animaux , Encéphale/effets des médicaments et des substances chimiques , Mâle , Neuro-immunomodulation/effets des médicaments et des substances chimiques , Neuroprotection/effets des médicaments et des substances chimiques , Rats , Rat Wistar , Sepsie/induit chimiquementRÉSUMÉ
BACKGROUND: Inflammatory/immunological serum markers are useful for the early detection of organ dysfunction, helping the diagnosis of sepsis. Although the detection of blood biomarkers is a standard practice, the use of noninvasive samples (eg saliva) would be beneficial. AIM: To investigate the saliva of hospitalized patients with and without sepsis and identify the levels of inflammatory markers such as C-reactive protein (CRP), procalcitonin (PCT), interleukin 6 (IL-6) and nitric oxide (NO). METHODS: Saliva samples were collected from 26 patients in intensive care unit with diagnosis of sepsis and from 26 without sepsis (control). The levels of CRP were determined by using latex agglutination test, whereas those of procalcitonin and IL-6 by ELISA and NO by the Griess reaction. RESULTS: Of 26 patients with sepsis, 14 were males (54%) with a mean age of 63.81 ± 3.48 years. The control group had the same distribution for gender, with mean age 65.04 ± 4.07 years. Sepsis group showed higher salivary concentrations of CRP, PCT, IL-6 and NO, with only levels of IL-6 being statistically different (P = .0001). CONCLUSIONS: Patients with sepsis had significantly higher levels of IL-6 in their saliva, suggesting that this biological sample could be useful in the diagnosis of this condition.
Sujet(s)
Protéine C-réactive/analyse , Interleukine-6/analyse , Monoxyde d'azote/analyse , Procalcitonine/analyse , Salive/composition chimique , Sepsie/diagnostic , Sujet âgé , Protéine C-réactive/immunologie , Études cas-témoins , Test ELISA , Éthylènediamines , Femelle , Hospitalisation , Humains , Interleukine-6/immunologie , Tests au latex , Mâle , Adulte d'âge moyen , Monoxyde d'azote/immunologie , Procalcitonine/immunologie , Sepsie/immunologie , SulfamidesRÉSUMÉ
Sepsis is a leading cause of morbidity and mortality in intensive care units. Previously, we identified Protein Kinase C-delta (PKCδ) as an important regulator of the inflammatory response in sepsis. An important issue in development of anti-inflammatory therapeutics is the risk of immunosuppression and inability to effectively clear pathogens. In this study, we investigated whether PKCδ inhibition prevented organ dysfunction and improved survival without compromising pathogen clearance. Sprague Dawley rats underwent sham surgery or cecal ligation and puncture (CLP) to induce sepsis. Post-surgery, PBS or a PKCδ inhibitor (200µg/kg) was administered intra-tracheally (IT). At 24 hours post-CLP, there was evidence of lung and kidney dysfunction. PKCδ inhibition decreased leukocyte influx in these organs, decreased endothelial permeability, improved gas exchange, and reduced blood urea nitrogen/creatinine ratios indicating organ protection. PKCδ inhibition significantly decreased bacterial levels in the peritoneal cavity, spleen and blood but did not exhibit direct bactericidal properties. Peritoneal chemokine levels, neutrophil numbers, or macrophage phenotypes were not altered by PKCδ inhibition. Peritoneal macrophages isolated from PKCδ inhibitor-treated septic rats demonstrated increased bacterial phagocytosis. Importantly, PKCδ inhibition increased survival. Thus, PKCδ inhibition improved survival and improved survival was associated with increased phagocytic activity, enhanced pathogen clearance, and decreased organ injury.
Sujet(s)
Bactéries/immunologie , Antienzymes/pharmacologie , Macrophages péritonéaux , Granulocytes neutrophiles , Protein kinase C-delta/antagonistes et inhibiteurs , Sepsie , Animaux , Chimiokines , Macrophages péritonéaux/immunologie , Macrophages péritonéaux/anatomopathologie , Mâle , Granulocytes neutrophiles/immunologie , Granulocytes neutrophiles/anatomopathologie , Phagocytose/effets des médicaments et des substances chimiques , Protein kinase C-delta/immunologie , Rats , Rat Sprague-Dawley , Sepsie/traitement médicamenteux , Sepsie/immunologie , Sepsie/microbiologie , Sepsie/anatomopathologieRÉSUMÉ
(-)-α-Bisabolol (BISA) is an unsaturated monocyclic sesquiterpenes compound, mainly found in the essential oil of chamomile (Matricaria chamomilla). It has been reported that this compound has several biological activities, but there are few studies evaluating the activity of this compound in the systemic inflammatory response in infectious processes. The aim of this study was to evaluate the effect of BISA on the inflammatory response and survival rate in a systemic infection model, and in vitro neutrophils phagocytic activity. BISA at concentration of 3, 10, 30, and 90 µg/ml did not presented in vitro cytotoxicity in MTT assay, and at concentrations of 1 and 3 µg/ml the BISA treatment increased in vitro phagocytic neutrophil activity. For the inflammatory response study, we verified the BISA treatment effect in a cecal ligation and puncture (CLP)-induced systemic infection model in mice; in this model, we demonstrate that BISA at dose of 100 mg/kg reduced the leukocyte recruitment in peritoneal cavity; at dose of 200 mg/kg, the NO concentration was increased in the peritoneal cavity. The bacteria CFU number was reduced in mice blood in the BISA treatment, at doses of 100 and 200 mg/kg. The BISA treatment at doses of 50 and 100 mg/kg increased the myeloperoxidase activity and reduction NO production in lung tissue of mice in CLP model. At dose of 100 mg/kg, the BISA treatment was able to reduce the mortality rate of mice submitted to CLP-induced sepsis and observed for 7 days. The results suggest an effect of BISA on inflammatory response, with activity on leukocyte chemotactic and NO production, in addition to increasing the survival rate of animals submitted to CLP model.
Sujet(s)
Anti-inflammatoires/pharmacologie , Inflammation/prévention et contrôle , Sesquiterpènes monocycliques/pharmacologie , Granulocytes neutrophiles/effets des médicaments et des substances chimiques , Phagocytose/effets des médicaments et des substances chimiques , Sepsie/traitement médicamenteux , Animaux , Charge bactérienne , Cellules cultivées , Chimiotaxie des leucocytes/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Femelle , Interactions hôte-pathogène , Inflammation/immunologie , Inflammation/métabolisme , Inflammation/microbiologie , Poumon/effets des médicaments et des substances chimiques , Poumon/immunologie , Poumon/métabolisme , Poumon/microbiologie , Souris de lignée C57BL , Granulocytes neutrophiles/immunologie , Granulocytes neutrophiles/métabolisme , Granulocytes neutrophiles/microbiologie , Monoxyde d'azote/métabolisme , Péritoine/effets des médicaments et des substances chimiques , Péritoine/immunologie , Péritoine/métabolisme , Péritoine/microbiologie , Sepsie/immunologie , Sepsie/métabolisme , Sepsie/microbiologieRÉSUMÉ
Sepsis is characterized by multiorgan system dysfunction that occurs because of infection. It is associated with high morbidity and mortality and is in need of improved therapeutic interventions. Neutrophils play a crucial role in sepsis, releasing neutrophil extracellular traps (NETs) composed of DNA complexed with histones and toxic antimicrobial proteins that ensnare pathogens, but also damage host tissues. At presentation, patients often have a significant NET burden contributing to the multiorgan damage. Therefore, interventions that inhibit NET release would likely be ineffective at preventing NET-based injury. Treatments that enhance NET degradation may liberate captured bacteria and toxic NET degradation products (NDPs) and likely be of limited therapeutic benefit as well. We propose that interventions that stabilize NETs and sequester NDPs may be protective in sepsis. We showed that platelet factor 4 (PF4), a platelet-associated chemokine, binds and compacts NETs, increasing their resistance to DNase I. We now show that PF4 increases NET-mediated bacterial capture, reduces the release of NDPs, and improves outcome in murine models of sepsis. A monoclonal antibody KKO which binds to PF4-NET complexes, further enhances DNase resistance. However, the Fc portion of this antibody activates the immune response and increases thrombotic risk, negating any protective effects in sepsis. Therefore, we developed an Fc-modified KKO that does not induce these negative outcomes. Treatment with this antibody augmented the effects of PF4, decreasing NDP release and bacterial dissemination and increasing survival in murine sepsis models, supporting a novel NET-targeting approach to improve outcomes in sepsis.