Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Br J Pharmacol ; 181(8): 1308-1323, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37990806

ABSTRACT

BACKGROUND AND PURPOSE: Sepsis-surviving adult individuals commonly develop immunosuppression and increased susceptibility to secondary infections, an outcome mediated by the axis IL-33/ILC2s/M2 macrophages/Tregs. Nonetheless, the long-term immune consequences of paediatric sepsis are indeterminate. We sought to investigate the role of age in the genesis of immunosuppression following sepsis. EXPERIMENTAL APPROACH: Here, we compared the frequency of Tregs, the activation of the IL-33/ILC2s axis in M2 macrophages and the DNA methylation of epithelial lung cells from post-septic infant and adult mice. Likewise, sepsis-surviving mice were inoculated intranasally with Pseudomonas aeruginosa or by subcutaneous inoculation of the B16 melanoma cell line. Finally, blood samples from sepsis-surviving patients were collected and the concentration of IL-33 and Tregs frequency were assessed. KEY RESULTS: In contrast to 6-week-old mice, 2-week-old mice were resistant to secondary infection and did not show impairment in tumour controls upon melanoma challenge. Mechanistically, increased IL-33 levels, Tregs expansion, and activation of ILC2s and M2-macrophages were observed in 6-week-old but not 2-week-old post-septic mice. Moreover, impaired IL-33 production in 2-week-old post-septic mice was associated with increased DNA methylation in lung epithelial cells. Notably, IL-33 treatment boosted the expansion of Tregs and induced immunosuppression in 2-week-old mice. Clinically, adults but not paediatric post-septic patients exhibited higher counts of Tregs and seral IL-33 levels. CONCLUSION AND IMPLICATIONS: These findings demonstrate a crucial and age-dependent role for IL-33 in post-sepsis immunosuppression. Thus, a better understanding of this process may lead to differential treatments for adult and paediatric sepsis.


Subject(s)
Interleukin-33 , Sepsis , Humans , Mice , Animals , Child , Immunity, Innate , Lymphocytes/metabolism , Lymphocytes/pathology , Immunosuppression Therapy
2.
Blood ; 138(25): 2702-2713, 2021 12 23.
Article in English | MEDLINE | ID: mdl-34407544

ABSTRACT

Multiple organ dysfunction is the most severe outcome of sepsis progression and is highly correlated with a worse prognosis. Excessive neutrophil extracellular traps (NETs) are critical players in the development of organ failure during sepsis. Therefore, interventions targeting NET release would likely effectively prevent NET-based organ injury associated with this disease. Herein, we demonstrate that the pore-forming protein gasdermin D (GSDMD) is active in neutrophils from septic humans and mice and plays a crucial role in NET release. Inhibition of GSDMD with disulfiram or genic deletion abrogated NET formation, reducing multiple organ dysfunction and sepsis lethality. Mechanistically, we demonstrate that during sepsis, activation of the caspase-11/GSDMD pathway controls NET release by neutrophils during sepsis. In summary, our findings uncover a novel therapeutic use for disulfiram and suggest that GSDMD is a therapeutic target to improve sepsis treatment.


Subject(s)
Extracellular Traps/genetics , Gene Deletion , Intracellular Signaling Peptides and Proteins/genetics , Multiple Organ Failure/genetics , Phosphate-Binding Proteins/genetics , Sepsis/genetics , Acetaldehyde Dehydrogenase Inhibitors/therapeutic use , Adoptive Transfer , Aged , Animals , Cells, Cultured , Disulfiram/therapeutic use , Female , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Male , Mice, Inbred C57BL , Middle Aged , Multiple Organ Failure/pathology , Multiple Organ Failure/therapy , Phosphate-Binding Proteins/antagonists & inhibitors , Sepsis/pathology , Sepsis/therapy
3.
Crit Care ; 23(1): 113, 2019 Apr 08.
Article in English | MEDLINE | ID: mdl-30961634

ABSTRACT

BACKGROUND: Neutrophil extracellular traps (NETs) are innate defense mechanisms that are also implicated in the pathogenesis of organ dysfunction. However, the role of NETs in pediatric sepsis is unknown. METHODS: Infant (2 weeks old) and adult (6 weeks old) mice were submitted to sepsis by intraperitoneal (i.p.) injection of bacteria suspension or lipopolysaccharide (LPS). Neutrophil infiltration, bacteremia, organ injury, and concentrations of cytokine, NETs, and DNase in the plasma were measured. Production of reactive oxygen and nitrogen species and release of NETs by neutrophils were also evaluated. To investigate the functional role of NETs, mice undergoing sepsis were treated with antibiotic plus rhDNase and the survival, organ injury, and levels of inflammatory markers and NETs were determined. Blood samples from pediatric and adult sepsis patients were collected and the concentrations of NETs measured. RESULTS: Infant C57BL/6 mice subjected to sepsis or LPS-induced endotoxemia produced significantly higher levels of NETs than the adult mice. Moreover, compared to that of the adult mice, this outcome was accompanied by increased organ injury and production of inflammatory cytokines. The increased NETs were associated with elevated expression of Padi4 and histone H3 citrullination in the neutrophils. Furthermore, treatment of infant septic mice with rhDNase or a PAD-4 inhibitor markedly attenuated sepsis. Importantly, pediatric septic patients had high levels of NETs, and the severity of pediatric sepsis was positively correlated with the level of NETs. CONCLUSION: This study reveals a hitherto unrecognized mechanism of pediatric sepsis susceptibility and suggests that NETs represents a potential target to improve clinical outcomes of sepsis.


Subject(s)
Extracellular Traps/microbiology , Sepsis/therapy , Animals , Bacterial Load/methods , Brazil , Disease Models, Animal , Mice , Mice, Inbred C57BL/blood , Mice, Inbred C57BL/microbiology , Multiple Organ Failure/etiology , Multiple Organ Failure/pathology , Sepsis/mortality , Sepsis/pathology
4.
J Infect Dis ; 219(12): 2015-2025, 2019 05 24.
Article in English | MEDLINE | ID: mdl-30715407

ABSTRACT

Rocio virus (ROCV) is a highly neuropathogenic mosquito-transmitted flavivirus responsible for an unprecedented outbreak of human encephalitis during 1975-1976 in Sao Paulo State, Brazil. Previous studies have shown an increased number of inflammatory macrophages in the central nervous system (CNS) of ROCV-infected mice, implying a role for macrophages in the pathogenesis of ROCV. Here, we show that ROCV infection results in increased expression of CCL2 in the blood and in infiltration of macrophages into the brain. Moreover, we show, using CCR2 knockout mice, that CCR2 expression is essential for macrophage infiltration in the brain during ROCV infection and that the lack of CCR2 results in increased disease severity and mortality. Thus, our findings show the protective role of CCR2-mediated infiltration of macrophages in the brain during ROCV infection.


Subject(s)
Encephalitis/metabolism , Flavivirus Infections/metabolism , Flavivirus/pathogenicity , Macrophages/metabolism , Receptors, CCR2/metabolism , Animals , Brain , Brazil , Encephalitis/virology , Female , Flavivirus Infections/virology , Macrophages/virology , Mice , Mice, Inbred C57BL , Mice, Knockout
5.
Front Immunol ; 10: 3108, 2019.
Article in English | MEDLINE | ID: mdl-32082301

ABSTRACT

The Chikungunya virus (CHIKV) is a re-emerging arbovirus, in which its infection causes a febrile illness also commonly associated with severe joint pain and myalgia. Although the immune response to CHIKV has been studied, a better understanding of the virus-host interaction mechanisms may lead to more effective therapeutic interventions. In this context, neutrophil extracellular traps (NETs) have been described as a key mediator involved in the control of many pathogens, including several bacteria and viruses, but no reports of this important protective mechanism were documented during CHIKV infection. Here we demonstrate that the experimental infection of mouse-isolated neutrophils with CHIKV resulted in NETosis (NETs release) through a mechanism dependent on TLR7 activation and reactive oxygen species generation. In vitro, mouse-isolated neutrophils stimulated with phorbol 12-myristate 13-acetate release NETs that once incubated with CHIKV, resulting in further virus capture and neutralization. In vivo, NETs inhibition by the treatment of the mice with DNase resulted in the enhanced susceptibility of IFNAR-/- mice to CHIKV experimental acute infection. Lastly, by accessing the levels of MPO-DNA complex on the acutely CHIKV-infected patients, we found a correlation between the levels of NETs and the viral load in the blood, suggesting that NETs are also released in natural human infection cases. Altogether our findings characterize NETosis as a contributing natural process to control CHIKV acute infection, presenting an antiviral effect that helps to control systemic virus levels.


Subject(s)
Chikungunya Fever/immunology , Chikungunya Fever/virology , Chikungunya virus/immunology , Extracellular Traps/immunology , Host-Pathogen Interactions/immunology , Neutrophils/immunology , Animals , Biomarkers , Cell Line , Chikungunya Fever/genetics , Disease Models, Animal , Disease Susceptibility , Extracellular Traps/genetics , Host-Pathogen Interactions/genetics , Immunity, Innate , Membrane Glycoproteins , Mice , Mice, Knockout , Neutralization Tests , Neutrophils/metabolism , Reactive Oxygen Species/metabolism , Toll-Like Receptor 7 , Viral Load , Virus Replication , Zika Virus/immunology
6.
Cancer Res ; 78(20): 5891-5900, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30104241

ABSTRACT

Paclitaxel is an antineoplastic agent widely used to treat several solid tumor types. The primary mechanism of action of paclitaxel is based on microtubule stabilization inducing cell-cycle arrest. Here, we use several tumor models to show that paclitaxel not only induces tumor cell-cycle arrest, but also promotes antitumor immunity. In vitro, paclitaxel reprogrammed M2-polarized macrophages to the M1-like phenotype in a TLR4-dependent manner, similarly to LPS. Paclitaxel also modulated the tumor-associated macrophage (TAM) profile in mouse models of breast and melanoma tumors; gene expression analysis showed that paclitaxel altered the M2-like signature of TAMs toward an M1-like profile. In mice selectively lacking TLR4 on myeloid cells, for example, macrophages (LysM-Cre+/-/TLR4fl/fl), the antitumor effect of paclitaxel was attenuated. Gene expression analysis of tumor samples from patients with ovarian cancer before and after treatment with paclitaxel detected an enrichment of genes linked to the M1 macrophage activation profile (IFNγ-stimulated macrophages). These findings indicate that paclitaxel skews TAMs toward an immunocompetent profile via TLR4, which might contribute to the antitumor effect of paclitaxel and provide a rationale for new combination regimens comprising paclitaxel and immunotherapies as an anticancer treatment.Significance: This study provides new evidence that the antitumor effect of paclitaxel occurs in part via reactivation of the immune response against cancer, guiding tumor-associated macrophages toward the M1-like antitumor phenotype.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/20/5891/F1.large.jpg Cancer Res; 78(20); 5891-900. ©2018 AACR See related commentary by Garassino et al., p. 5729.


Subject(s)
Macrophages/metabolism , Neoplasms/pathology , Paclitaxel/pharmacology , Toll-Like Receptor 4/metabolism , Animals , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Immune System , Immunotherapy , Macrophage Activation , Melanoma/drug therapy , Melanoma/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NF-kappa B/metabolism , Neoplasms/drug therapy , Oligonucleotide Array Sequence Analysis , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Signal Transduction , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology
7.
PLoS One ; 13(6): e0199071, 2018.
Article in English | MEDLINE | ID: mdl-29897990

ABSTRACT

Rocio virus (ROCV) was the causative agent of an unprecedented outbreak of encephalitis during the 1970s in the Vale do Ribeira, Sao Paulo State, in the Southeast region of Brazil. Surprisingly, no further cases of ROCV infection were identified after this outbreak; however, serological surveys have suggested the circulation of ROCV among humans and animals in different regions of Brazil. Cross-protective immunity among flaviviruses is well documented; consequently, immunity induced by infections with other flaviviruses endemic to Brazil could potentially be responsible for the lack of ROCV infections. Herein, we evaluated the cross-protection mediated by other flaviviruses against ROCV infection using an experimental C57BL/6 mouse model. Cross-protection against ROCV infection was observed when animals had prior exposure to Ilheus virus or Saint Louis encephalitis virus, suggesting that cross-reactive anti-flavivirus antibodies may limit ROCV disease outbreaks.


Subject(s)
Encephalitis Virus, St. Louis/immunology , Flavivirus Infections/prevention & control , Flavivirus/pathogenicity , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Disease Susceptibility , Encephalitis Virus, St. Louis/pathogenicity , Evolution, Molecular , Female , Flavivirus Infections/immunology , Flavivirus Infections/mortality , Flavivirus Infections/veterinary , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Survival Rate
8.
Nat Commun ; 8: 14919, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28374774

ABSTRACT

Patients who survive sepsis can develop long-term immune dysfunction, with expansion of the regulatory T (Treg) cell population. However, how Treg cells proliferate in these patients is not clear. Here we show that IL-33 has a major function in the induction of this immunosuppression. Mice deficient in ST2 (IL-33R) develop attenuated immunosuppression in cases that survive sepsis, whereas treatment of naive wild-type mice with IL-33 induces immunosuppression. IL-33, released during tissue injury in sepsis, activates type 2 innate lymphoid cells, which promote polarization of M2 macrophages, thereby enhancing expansion of the Treg cell population via IL-10. Moreover, sepsis-surviving patients have more Treg cells, IL-33 and IL-10 in their peripheral blood. Our study suggests that targeting IL-33 may be an effective treatment for sepsis-induced immunosuppression.


Subject(s)
Immune Tolerance/immunology , Interleukin-33/immunology , Sepsis/immunology , T-Lymphocytes, Regulatory/immunology , Aged , Animals , Female , Humans , Immune Tolerance/genetics , Interleukin-1 Receptor-Like 1 Protein/deficiency , Interleukin-1 Receptor-Like 1 Protein/genetics , Interleukin-1 Receptor-Like 1 Protein/immunology , Interleukin-10/immunology , Interleukin-10/metabolism , Interleukin-33/metabolism , Macrophages/immunology , Macrophages/metabolism , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Sepsis/genetics , Sepsis/metabolism , T-Lymphocytes, Regulatory/metabolism
9.
Pharmacol Res ; 117: 1-8, 2017 03.
Article in English | MEDLINE | ID: mdl-27979692

ABSTRACT

Sepsis is one of the main causes of mortality in hospitalized patients. Despite the recent technical advances and the development of novel generation of antibiotics, severe sepsis remains a major clinical and scientific challenge in modern medicine. Unsuccessful efforts have been dedicated to the search of therapeutic options to treat the deleterious inflammatory components of sepsis. Recent findings on neuronal networks controlling immunity raised expectations for novel therapeutic strategies to promote the regulation of sterile inflammation, such as autoimmune diseases. Interesting studies have dissected the anatomical constituents of the so-called "cholinergic anti-inflammatory pathway", suggesting that electrical vagus nerve stimulation and pharmacological activation of beta-2 adrenergic and alpha-7 nicotinic receptors could be alternative strategies for improving inflammatory conditions. However, the literature on infectious diseases, such as sepsis, is still controversial and, therefore, the real therapeutic potential of this neuroimmune pathway is not well defined. In this review, we will discuss the beneficial and detrimental effects of neural manipulation in sepsis, which depend on the multiple variables of the immune system and the nature of the infection. These observations suggest future critical studies to validate the clinical implications of vagal parasympathetic signaling in sepsis treatment.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Cholinergic Agents/pharmacology , Cholinergic Agents/therapeutic use , Sepsis/drug therapy , Animals , Humans , Immune System/drug effects , Inflammation/drug therapy , Signal Transduction/drug effects , Vagus Nerve/drug effects
10.
J Bone Miner Res ; 32(5): 974-984, 2017 May.
Article in English | MEDLINE | ID: mdl-28029186

ABSTRACT

Chemerin is an adipokine that regulates adipogenesis and metabolic functions of mature adipocytes mainly through the activation of chemokine-like receptor 1 (CMKLR1). Elevated levels of chemerin have been found in individuals with obesity, type 2 diabetes, and osteoporosis. This adipokine was identified as an inflammatory and metabolic syndrome marker. Considering that the association between metabolic syndrome and bone health remains unclear, the present study aimed to clarify the role of chemerin in the pathophysiology of bone loss induced by dyslipidemia, particularly modulating osteoclastogenesis. In vitro analyses showed a downregulation of CMKLR1 at the early stage of differentiation and a gradual increase at late stages. Strikingly, chemerin did not modify osteoclast differentiation markers or osteoclast formation; however, it increased the actin-ring formation and bone resorption activity in mature osteoclasts. The increased bone resorption activity induced by chemerin was effectively inhibited by CMKLR1 antagonist (CCX832). Chemerin boosting mature osteoclast activity involves ERK5 phosphorylation. Moreover, two models of dyslipidemia (high-fat diet [HFD]-treated C57/BL6 and db/db mice) exhibited significantly increased level of chemerin in the serum and gingival tissue. Morphometric analysis showed that HFD-treated and db/db mice exhibited increased alveolar bone loss compared to respective control mice, which was associated with an up-regulation of chemerin, CMKLR1 and cathepsin K mRNA expression in the gingival tissue. The treatment of db/db mice with CCX832 effectively inhibited bone loss. Antagonism of chemerin receptor also inhibited the expression of cathepsin K in the gingival tissue. Our results show that chemerin not only increases osteoclasts activity in vitro, but also that increased level of chemerin in dyslipidemic mice plays a critical role in bone homeostasis. © 2016 American Society for Bone and Mineral Research.


Subject(s)
Alveolar Bone Loss/metabolism , Chemokines/metabolism , Dyslipidemias/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Osteoclasts/metabolism , Alveolar Bone Loss/chemically induced , Alveolar Bone Loss/pathology , Animals , Dietary Fats/adverse effects , Dietary Fats/pharmacology , Dyslipidemias/chemically induced , Dyslipidemias/pathology , Male , Mice , Osteoclasts/pathology , Receptors, Chemokine , Receptors, G-Protein-Coupled/metabolism
11.
Sci Rep ; 6: 36401, 2016 11 07.
Article in English | MEDLINE | ID: mdl-27819273

ABSTRACT

Isoflavonoids have been largely studied due to their distinct biological activities identified thus far. Herein, we evaluated the activity of neovestitol, an isoflavonoid isolated from Brazilian red propolis, in acute and chronic inflammation. As for acute inflammation, we found that neovestitol reduced neutrophil migration, leukocyte rolling and adhesion, as well as expression of ICAM-1 in the mesenteric microcirculation during lipopolysaccharide-induced acute peritonitis. No changes were observed in the levels of TNF-α, CXCL1/KC and CXCL2/MIP-2 upon pretreatment with neovestitol. The administration of an inducible nitric oxide synthase (iNOS) inhibitor abolished the inhibitory effects of neovestitol in neutrophil migration and ICAM-1 expression. Nitrite levels increased upon treatment with neovestitol. No effects of neovestitol were observed on the chemotaxis of neutrophils in vitro. As for chronic inflammation, neovestitol also reduced the clinical score and joint damage in a collagen-induced arthritis model. There was no change in the frequency of IL-17-producing TCD4+ cells. In addition, pretreatment with neovestitol reduced the levels of IL-6. These results demonstrate a potential anti-inflammatory activity of neovestitol, which may be useful for therapeutic purposes and/or as a nutraceutical.


Subject(s)
Arthritis, Experimental/prevention & control , Flavonoids/therapeutic use , Interleukin-6/metabolism , Nitric Oxide/metabolism , Peritonitis/prevention & control , Propolis/chemistry , Acute Disease , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Arthritis, Experimental/etiology , Brazil , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Cell Line , Cell Movement/drug effects , Cell Survival/drug effects , Chronic Disease , Cytokines/metabolism , Flavonoids/chemistry , Flavonoids/pharmacology , Guanidines/pharmacology , Lipopolysaccharides/toxicity , Mesenteric Veins/drug effects , Mesenteric Veins/metabolism , Mesenteric Veins/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Neutrophils/cytology , Neutrophils/drug effects , Neutrophils/immunology , Peritonitis/etiology , Propolis/metabolism
12.
J Nat Prod ; 79(7): 1828-33, 2016 07 22.
Article in English | MEDLINE | ID: mdl-27367493

ABSTRACT

Chemical compounds belonging to the class of coumarins have promising anti-inflammatory potential. Cinnamoyloxy-mammeisin (CNM) is a 4-phenylcoumarin that can be isolated from Brazilian geopropolis. To our knowledge, its anti-inflammatory activity has never been studied. Therefore, the present study investigated the anti-inflammatory activity of CNM and elucidated its mechanism of action on isolated macrophages. Pretreatment with CNM reduced neutrophil migration into the peritoneal and joint cavity of mice. Likewise, CNM reduced the in vitro and in vivo release of TNF-α and CXCL2/MIP-2. Regarding the possible molecular mechanism of action, CNM reduced the phosphorylation of proteins ERK 1/2, JNK, p38 MAPK, and AP-1 (subunit c-jun) in PG-stimulated macrophages. Pretreatment with CNM also reduced NF-κB activation in RAW 264.7 macrophages stably expressing the NF-κB-luciferase reporter gene. On the other hand, it did not alter IκBα degradation or nuclear translocation of p65. Thus, the results of this study demonstrate promising anti-inflammatory activity of CNM and provide an explanation of its mechanism of action in macrophages via inhibition of MAPK signaling, AP-1, and NF-κB.


Subject(s)
Anti-Inflammatory Agents/isolation & purification , Anti-Inflammatory Agents/pharmacology , Coumarins/isolation & purification , Coumarins/pharmacology , Animals , Anti-Inflammatory Agents/chemistry , Brazil , Coumarins/chemistry , Cyclooxygenase 2/metabolism , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Mice , Molecular Structure , NF-kappa B/antagonists & inhibitors , Nitric Oxide Synthase Type II/antagonists & inhibitors , Signal Transduction/drug effects , Transcription Factor AP-1 , Tumor Necrosis Factor-alpha/pharmacology , eIF-2 Kinase/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
13.
J Neuroinflammation ; 13(1): 159, 2016 06 22.
Article in English | MEDLINE | ID: mdl-27334012

ABSTRACT

BACKGROUND: Viral encephalitis is a common cause of lethal infections in humans, and several different viruses are documented to be responsible. Rocio virus is a flavivirus that causes a severe lethal encephalitis syndrome in humans and also mice, providing an interesting model to study the CNS compartmentalized immune response. Interleukin 33 (IL-33), a member of the IL-1 family, is an immunomodulatory cytokine that is highly expressed in the CNS. However, the role of IL-33 on viral encephalitis remains unclear. Therefore, we aimed to explore how the IL-33/ST2 axis regulates the local immune response during Rocio virus infection. METHODS: Wild-type (WT), ST2 (ST2(-/-)), and nitric oxide synthase-deficient mice (iNOS(-/-)) and Stat6 (Stat6(-/-))-deficient mice were infected with different concentrations of the Rocio virus by intraperitoneal route, the cytokine mRNA level in CNS was analyzed by qPCR, and cellular immunophenotyping was performed on infected mice by the flow cytometry of isolated CNS mononuclear cells. RESULTS: We have shown that the mRNA expression of IL-33 and ST2 receptors is increased in the CNS of Rocio virus-infected WT mice and that ST2(-/-) mice showed increased susceptibility to infection. ST2 deficiency was correlated with increased tissue pathology, cellular infiltration, and tumor necrosis factor alpha (TNF-α) and interferon-gamma (IFN-γ) mRNA levels and higher viral load in the CNS, compared with wild-type mice. The increased Th1 cytokine levels released in the CNS acted on infiltrating macrophages, as evidenced by flow cytometry characterization of cellular infiltrates, inducing the expression of iNOS, contributing to brain injury. Moreover, iNOS(-/-) mice were more resistant to Rocio virus encephalitis, presenting a lower clinical score and reduced mortality rate, despite the increased tissue pathology. CONCLUSIONS: We provide evidences of a specific role for IL-33 receptor signaling in nitric oxide induction through local IFN-γ modulation, suggesting that nitric oxide overproduction might have an important role in the progression of experimental viral encephalitis.


Subject(s)
Central Nervous System , Encephalitis, Viral/pathology , Interleukin-33/metabolism , Nitric Oxide Synthase Type II/metabolism , Animals , Antigens, Differentiation/metabolism , Central Nervous System/immunology , Central Nervous System/pathology , Central Nervous System/virology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Down-Regulation/genetics , Female , Flaviviridae Infections/pathology , Flow Cytometry , Interleukin-1 Receptor-Like 1 Protein/deficiency , Interleukin-1 Receptor-Like 1 Protein/genetics , Interleukin-33/genetics , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Nitric Oxide Synthase Type II/genetics , RNA, Messenger/metabolism , STAT6 Transcription Factor/deficiency , STAT6 Transcription Factor/genetics , Signal Transduction/physiology
14.
J Nat Prod ; 79(4): 954-60, 2016 Apr 22.
Article in English | MEDLINE | ID: mdl-26938776

ABSTRACT

Vestitol is an isoflavonoid isolated from Brazilian red propolis with potential anti-inflammatory activity. This study investigated the mechanism of action of vestitol on the modulation of neutrophil migration in the inflammatory process. Pre-treatment with vestitol at 1, 3, or 10 mg/kg reduced LPS- or mBSA-induced neutrophil migration and the release of CXCL1/KC and CXCL2/MIP-2 in vivo. Likewise, pre-treatment with vestitol at 1, 3, or 10 µM reduced the levels of CXCL1/KC and CXCL2/MIP-2 in macrophage supernatants in vitro. Moreover, the administration of vestitol (10 mg/kg) reduced leukocyte rolling and adherence in the mesenteric microcirculation of mice. The pre-treatment with vestitol (10 mg/kg) in iNOS(-/-) mice did not block its activity concerning neutrophil migration. With regard to the activity of vestitol on neutrophils isolated from the bone marrow of mice, there was a reduction on the chemotaxis of CXCL2/MIP-2 or LTB4-induced neutrophils and on calcium influx after pre-treatment with the compound at 3 or 10 µM. There was no change in CXCR2 expression by neutrophils treated with vestitol at 10 µM. These findings demonstrate that vestitol is a promising novel anti-inflammatory agent.


Subject(s)
Anti-Inflammatory Agents/isolation & purification , Flavonoids/isolation & purification , Neutrophils/drug effects , Propolis/chemistry , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Brazil , Chemokine CXCL1 , Flavonoids/chemistry , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred BALB C , Molecular Structure , Neutrophil Infiltration , Nitric Oxide Synthase Type II/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...