Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Cells ; 11(17)2022 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-36078125

RESUMO

Chikungunya (CHIKV) is an arthritogenic alphavirus that causes a self-limiting disease usually accompanied by joint pain and/or polyarthralgia with disabling characteristics. Immune responses developed during the acute phase of CHIKV infection determine the rate of disease progression and resolution. Annexin A1 (AnxA1) is involved in both initiating inflammation and preventing over-response, being essential for a balanced end of inflammation. In this study, we investigated the role of the AnxA1-FPR2/ALX pathway during CHIKV infection. Genetic deletion of AnxA1 or its receptor enhanced inflammatory responses driven by CHIKV. These knockout mice showed increased neutrophil accumulation and augmented tissue damage at the site of infection compared with control mice. Conversely, treatment of wild-type animals with the AnxA1 mimetic peptide (Ac2-26) reduced neutrophil accumulation, decreased local concentration of inflammatory mediators and diminished mechanical hypernociception and paw edema induced by CHIKV-infection. Alterations in viral load were mild both in genetic deletion or with treatment. Combined, our data suggest that the AnxA1-FPR2/ALX pathway is a potential therapeutic strategy to control CHIKV-induced acute inflammation and polyarthralgia.


Assuntos
Febre de Chikungunya , Inflamação , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Anexina A1/genética , Anexina A1/metabolismo , Artralgia , Febre de Chikungunya/metabolismo , Inflamação/metabolismo , Camundongos , Camundongos Knockout , Receptores de Formil Peptídeo/metabolismo
2.
ASN Neuro ; 14: 17590914221121257, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36017573

RESUMO

SUMMARY STATEMENT: In utero exposure to ZIKV leads to decreased number of neurons in adult mice. Female mice exposed to ZIKV in utero exhibit lower levels of BDNF, a decrease in synaptic markers, memory deficits, and risk-taking behavior during adulthood.


Assuntos
Infecção por Zika virus , Zika virus , Animais , Feminino , Masculino , Transtornos da Memória/etiologia , Camundongos , Neurônios , Infecção por Zika virus/complicações
3.
Stem Cell Rev Rep ; 17(5): 1874-1888, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34003465

RESUMO

Multiple infectious diseases lead to impaired lung function. Revealing the cellular mechanisms involved in this impairment is crucial for the understanding of how the lungs shift from a physiologic to a pathologic state in each specific condition. In this context, we explored the pathogenesis of Paracoccidioidomycosis, which affects pulmonary functioning. The presence of cells expressing Nestin-GFP has been reported in different tissues, and their roles as tissue-specific progenitors have been stablished in particular organs. Here, we explored how Nestin-GFP+ cells are affected after lung infection by Paracoccidioides brasiliensis, a model of lung granulomatous inflammation with fibrotic outcome. We used Nestin-GFP transgenic mice, parabiosis surgery, confocal microscopy and flow cytometry to investigate the participation of Nestin-GFP+ cells in Paracoccidioides brasiliensis pathogenesis. We revealed that these cells increase in the lungs post-Paracoccidioides brasiliensis infection, accumulating around granulomas. This increase was due mainly to Nestin-GPF+ cells derived from the blood circulation, not associated to blood vessels, that co-express markers suggestive of hematopoietic cells (Sca-1, CD45 and CXCR4). Therefore, our findings suggest that circulating Nestin-GFP+ cells participate in the Paracoccidioides brasiliensis pathogenesis in the lungs.


Assuntos
Pulmão , Animais , Camundongos , Nestina/genética , Paracoccidioides/genética
4.
Front Immunol ; 10: 1461, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31354697

RESUMO

Reperfusion of an ischemic tissue is the treatment of choice for several diseases, including myocardial infarction and stroke. However, reperfusion of an ischemic tissue causes injury, known as Ischemia and Reperfusion Injury (IRI), that limits the benefit of blood flow restoration. IRI also occurs during solid organ transplantation. During IRI, there is activation of the innate immune system, especially neutrophils, which contributes to the degree of injury. It has been shown that PTX3 can regulate multiple aspects of innate immunity and tissue inflammation during sterile injury, as observed during IRI. In humans, levels of PTX3 increase in blood and elevated levels associate with extent of IRI. In mice, there is also enhanced expression of PTX3 in tissues and plasma after IRI. In general, absence of PTX3, as seen in PTX3-deficient mice, results in worse outcome after IRI. On the contrary, increased expression of PTX3, as seen in PTX3 transgenic mice and after PTX3 administration, is associated with better outcome after IRI. The exception is the gut where PTX3 seems to have a clear deleterious role. Here, we discuss mechanisms by which PTX3 contributes to IRI and the potential of taming this system for the treatment of injuries associated with reperfusion of solid organs.


Assuntos
Proteína C-Reativa/metabolismo , Neutrófilos/imunologia , Traumatismo por Reperfusão/metabolismo , Componente Amiloide P Sérico/metabolismo , Animais , Proteína C-Reativa/genética , Regulação da Expressão Gênica , Humanos , Imunidade Inata , Camundongos , Camundongos Knockout , Especificidade de Órgãos , Componente Amiloide P Sérico/genética
5.
Viruses ; 11(6)2019 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-31212905

RESUMO

Zika virus (ZIKV) only induces mild symptoms in adults; however, it can cause congenital Zika syndrome (CZS), including microcephaly. Most of the knowledge on ZIKV pathogenesis was gained using immunocompromised mouse models, which do not fully recapitulate human pathology. Moreover, the study of the host immune response to ZIKV becomes challenging in these animals. Thus, the main goal of this study was to develop an immunocompetent mouse model to study the ZIKV spread and teratogeny. FVB/NJ immune competent dams were infected intravaginally with ZIKV during the early stage of pregnancy. We found that the placentae of most fetuses were positive for ZIKV, while the virus was detected in the brain of only about 42% of the embryos. To investigate the host immune response, we measured the expression of several inflammatory factors. Embryos from ZIKV-infected dams had an increased level of inflammatory factors, as compared to Mock. Next, we compared the gene expression levels in embryos from ZIKV-infected dams that were either negative or positive for ZIKV in the brain. The mRNA levels of viral response genes and cytokines were increased in both ZIKV-positive and negative brains. Interestingly, the levels of chemokines associated with microcephaly in humans, including CCL2 and CXCL10, specifically increased in embryos harboring ZIKV in the embryo brains.


Assuntos
Modelos Animais de Doenças , Interações Hospedeiro-Patógeno , Complicações Infecciosas na Gravidez/patologia , Infecção por Zika virus/patologia , Zika virus/patogenicidade , Animais , Encéfalo/virologia , Feminino , Perfilação da Expressão Gênica , Fatores Imunológicos/biossíntese , Camundongos , Placenta/virologia , Gravidez , Complicações Infecciosas na Gravidez/virologia
6.
EBioMedicine ; 44: 516-529, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31130472

RESUMO

BACKGROUND: Zika virus (ZIKV) infection during pregnancy may cause major congenital defects, including microcephaly, ocular, articular and muscle abnormalities, which are collectively defined as Congenital Zika Syndrome. Here, we performed an in-depth characterization of the effects of congenital ZIKV infection (CZI) in immunocompetent mice. METHODS: Pregnant dams were inoculated with ZIKV on embryonic day 5.5 in the presence or absence of a sub-neutralizing dose of a pan-flavivirus monoclonal antibody (4G2) to evaluate the potential role of antibody-dependent enhancement phenomenon (ADE) during short and long outcomes of CZI. FINDINGS: ZIKV infection induced maternal immune activation (MIA), which was associated with occurrence of foetal abnormalities and death. Therapeutic administration of AH-D antiviral peptide during the early stages of pregnancy prevented ZIKV replication and death of offspring. In the post-natal period, CZI was associated with a decrease in whole brain volume, ophthalmologic abnormalities, changes in testicular morphology, and disruption in bone microarchitecture. Some alterations were enhanced in the presence of 4G2 antibody. INTERPRETATION: Our results reveal that early maternal ZIKV infection causes several birth defects in immunocompetent mice, which can be potentiated by ADE phenomenon and are associated with MIA. Additionally, antiviral treatment with AH-D peptide may be beneficial during early maternal ZIKV infection. FUND: This work was supported by the Brazilian National Science Council (CNPq, Brazil), Minas Gerais Foundation for Science (FAPEMIG), Funding Authority for Studies and Projects (FINEP), Coordination of Superior Level Staff Improvement (CAPES), National Research Foundation of Singapore and Centre for Precision Biology at Nanyang Technological University.


Assuntos
Anticorpos Facilitadores/imunologia , Interações Hospedeiro-Patógeno/imunologia , Complicações Infecciosas na Gravidez , Infecção por Zika virus/imunologia , Infecção por Zika virus/virologia , Zika virus/fisiologia , Animais , Anticorpos Antivirais/imunologia , Antivirais/farmacologia , Osso e Ossos/diagnóstico por imagem , Osso e Ossos/patologia , Encéfalo/efeitos dos fármacos , Encéfalo/imunologia , Encéfalo/patologia , Encéfalo/virologia , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Peptídeos/farmacologia , Gravidez , Baço/efeitos dos fármacos , Baço/imunologia , Baço/patologia , Baço/virologia , Síndrome , Resultado do Tratamento , Carga Viral , Infecção por Zika virus/diagnóstico , Infecção por Zika virus/tratamento farmacológico
7.
PLoS Negl Trop Dis ; 13(5): e0007375, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31050676

RESUMO

BACKGROUND: The Mayaro virus (MAYV) is an endemic arbovirus in South American countries, where it is responsible for sporadic outbreaks of Mayaro fever. Clinical manifestations include fever, headache, ocular pain, rash, myalgia, and debilitating and persistent polyarthralgia. Understanding the mechanisms associated with MAYV-induced arthritis is of great importance due to the potential for its emergence, urbanization and dispersion to other regions. METHODS: 15-day old Balb/c mice were infected by two distinct pathways, below the forelimb and in the rear footpad. Animals were observed for a period of 21 days. During this time, they were monitored every 24 hours for disease signs, such as weight loss and muscle weakness. Histological damage in the muscles and joints was evaluated 3, 7, 10, 15 and 20 days post-infection. The cytokine profile in serum and muscles during MAYV infection was evaluated by flow cytometry at different post-infection times. For pain analysis, the animals were submitted to the von Frey test and titre in different organs was evaluated throughout the study to obtain viral kinetics. FINDINGS: Infection by two distinct pathways, below the forelimb and in the rear footpad, resulted in a homogeneous viral spread and the development of acute disease in animals. Clinical signs were observed such as ruffled fur, hunched posture, eye irritation and slight gait alteration. In the physical test, both groups presented loss of resistance, which was associated with histopathological damage, including myositis, arthritis, tenosynovitis and periostitis. The immune response was characterized by a strong inflammatory response mediated by the cytokines TNF-α, IL-6 and INF-γ and chemokine MCP-1, followed by the action of IL-10 and IL-4 cytokines. INTERPRETATION: The results showed that Balb/c mice represent a promising model to study mechanisms involved in MAYV pathogenesis and for future antiviral testing.


Assuntos
Infecções por Arbovirus/virologia , Arbovírus/fisiologia , Artrite/virologia , Modelos Animais de Doenças , Miosite/virologia , Animais , Arbovírus/genética , Arbovírus/isolamento & purificação , Feminino , Humanos , Interferon gama/genética , Interferon gama/imunologia , Interleucina-10/genética , Interleucina-10/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Masculino , Camundongos Endogâmicos BALB C , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
8.
Nat Mater ; 17(11): 971-977, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30349030

RESUMO

Zika virus is a mosquito-borne virus that is associated with neurodegenerative diseases, including Guillain-Barré syndrome1 and congenital Zika syndrome2. As Zika virus targets the nervous system, there is an urgent need to develop therapeutic strategies that inhibit Zika virus infection in the brain. Here, we have engineered a brain-penetrating peptide that works against Zika virus and other mosquito-borne viruses. We evaluated the therapeutic efficacy of the peptide in a lethal Zika virus mouse model exhibiting systemic and brain infection. Therapeutic treatment protected against mortality and markedly reduced clinical symptoms, viral loads and neuroinflammation, as well as mitigated microgliosis, neurodegeneration and brain damage. In addition to controlling systemic infection, the peptide crossed the blood-brain barrier to reduce viral loads in the brain and protected against Zika-virus-induced blood-brain barrier injury. Our findings demonstrate how engineering strategies can be applied to develop peptide therapeutics and support the potential of a brain-penetrating peptide to treat neurotropic viral infections.


Assuntos
Antivirais/uso terapêutico , Encéfalo/metabolismo , Peptídeos/uso terapêutico , Infecção por Zika virus/tratamento farmacológico , Animais , Antivirais/farmacocinética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Peptídeos/farmacocinética
9.
Immunology ; 155(4): 477-490, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30098206

RESUMO

The excessive inflammation often present in patients with severe dengue infection is considered both a hallmark of disease and a target for potential treatments. Interleukin-33 (IL-33) is a pleiotropic cytokine with pro-inflammatory effects whose role in dengue has not been fully elucidated. We demonstrate that IL-33 plays a disease-exacerbating role during experimental dengue infection in immunocompetent mice. Mice infected with dengue virus serotype 2 (DENV2) produced high levels of IL-33. DENV2-infected mice treated with recombinant IL-33 developed markedly more severe disease compared with untreated mice as assessed by mortality, granulocytosis, liver damage and pro-inflammatory cytokine production. Conversely, ST2-/- mice (deficient in IL-33 receptor) infected with DENV2 developed significantly less severe disease compared with wild-type mice. Furthermore, the increased disease severity and the accompanying pathology induced by IL-33 during dengue infection were reversed by the simultaneous treatment with a CXCR2 receptor antagonist (DF2156A). Together, these results indicate that IL-33 plays a disease-exacerbating role in experimental dengue infection, probably driven by CXCR2-expressing cells, leading to elevated pro-inflammatory response-mediated pathology. Our results also indicate that IL-33 is a potential therapeutic target for dengue infection.


Assuntos
Vírus da Dengue/imunologia , Interleucina-33/farmacologia , Receptores de Interleucina-8B/antagonistas & inibidores , Proteínas Recombinantes/farmacologia , Animais , Dengue/imunologia , Dengue/virologia , Progressão da Doença , Proteína 1 Semelhante a Receptor de Interleucina-1/deficiência , Proteína 1 Semelhante a Receptor de Interleucina-1/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Sulfonamidas/farmacologia
10.
Artigo em Inglês | MEDLINE | ID: mdl-29463546

RESUMO

The clinical pathogen Klebsiella pneumoniae is a relevant cause of nosocomial infections, and resistance to current treatment with carbapenem antibiotics is becoming a significant problem. Statins are inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) used for controlling plasma cholesterol levels. There is clinical evidence showing other effects of statins, including decrease of lung inflammation. In the current study, we show that pretreatment with atorvastatin markedly attenuated lung injury, which was correlated with a reduction in the cellular influx into the alveolar space and lungs and downmodulation of the production of proinflammatory mediators in the initial phase of infection in C57BL/6 mice with K. pneumoniae However, atorvastatin did not alter the number of bacteria in the lungs and blood of infected mice, despite decreasing local inflammatory response. Interestingly, mice that received combined treatment with atorvastatin and imipenem displayed better survival than mice treated with vehicle, atorvastatin, or imipenem alone. These findings suggest that atorvastatin could be an adjuvant in host-directed therapies for multidrug-resistant K. pneumoniae, based on its powerful pleiotropic immunomodulatory effects. Together with antimicrobial approaches, combination therapy with anti-inflammatory compounds could improve the efficiency of therapy during acute lung infections.


Assuntos
Antibacterianos/uso terapêutico , Atorvastatina/uso terapêutico , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Imipenem/uso terapêutico , Infecções por Klebsiella/tratamento farmacológico , Klebsiella pneumoniae/efeitos dos fármacos , Pneumonia Bacteriana/tratamento farmacológico , Animais , Carga Bacteriana/efeitos dos fármacos , Quimiocinas/análise , Infecções Comunitárias Adquiridas/tratamento farmacológico , Infecções Comunitárias Adquiridas/microbiologia , Farmacorresistência Bacteriana Múltipla , Quimioterapia Combinada , Feminino , Inflamação/tratamento farmacológico , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/imunologia , Fagocitose/efeitos dos fármacos , Fagocitose/imunologia , Pneumonia Bacteriana/microbiologia
11.
Front Immunol ; 8: 1016, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28878777

RESUMO

Zika virus (ZIKV) has recently caused a worldwide outbreak of infections associated with severe neurological complications, including microcephaly in infants born from infected mothers. ZIKV exhibits high neurotropism and promotes neuroinflammation and neuronal cell death. We have recently demonstrated that N-methyl-d-aspartate receptor (NMDAR) blockade by memantine prevents ZIKV-induced neuronal cell death. Here, we show that ZIKV induces apoptosis in a non-cell autonomous manner, triggering cell death of uninfected neurons by releasing cytotoxic factors. Neuronal cultures infected with ZIKV exhibit increased levels of tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), and glutamate. Moreover, infected neurons exhibit increased expression of GluN2B and augmented intracellular Ca2+ concentration. Blockade of GluN2B-containing NMDAR by ifenprodil normalizes Ca2+ levels and rescues neuronal cell death. Notably, TNF-α and IL-1ß blockade decreases ZIKV-induced Ca2+ flux through GluN2B-containing NMDARs and reduces neuronal cell death, indicating that these cytokines might contribute to NMDAR sensitization and neurotoxicity. In addition, ZIKV-infected cultures treated with ifenprodil exhibits increased activation of the neuroprotective pathway including extracellular signal-regulated kinase and cAMP response element-binding protein, which may underlie ifenprodil-mediated neuroprotection. Together, our data shed some light on the neurotoxic mechanisms triggered by ZIKV and begin to elucidate how GluN2B-containing NMDAR blockade can prevent neurotoxicity.

12.
mBio ; 8(2)2017 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-28442607

RESUMO

Zika virus (ZIKV) infection is a global health emergency that causes significant neurodegeneration. Neurodegenerative processes may be exacerbated by N-methyl-d-aspartate receptor (NMDAR)-dependent neuronal excitoxicity. Here, we have exploited the hypothesis that ZIKV-induced neurodegeneration can be rescued by blocking NMDA overstimulation with memantine. Our results show that ZIKV actively replicates in primary neurons and that virus replication is directly associated with massive neuronal cell death. Interestingly, treatment with memantine or other NMDAR blockers, including dizocilpine (MK-801), agmatine sulfate, or ifenprodil, prevents neuronal death without interfering with the ability of ZIKV to replicate in these cells. Moreover, in vivo experiments demonstrate that therapeutic memantine treatment prevents the increase of intraocular pressure (IOP) induced by infection and massively reduces neurodegeneration and microgliosis in the brain of infected mice. Our results indicate that the blockade of NMDARs by memantine provides potent neuroprotective effects against ZIKV-induced neuronal damage, suggesting it could be a viable treatment for patients at risk for ZIKV infection-induced neurodegeneration.IMPORTANCE Zika virus (ZIKV) infection is a global health emergency associated with serious neurological complications, including microcephaly and Guillain-Barré syndrome. Infection of experimental animals with ZIKV causes significant neuronal damage and microgliosis. Treatment with drugs that block NMDARs prevented neuronal damage both in vitro and in vivo These results suggest that overactivation of NMDARs contributes significantly to the neuronal damage induced by ZIKV infection, and this is amenable to inhibition by drug treatment.


Assuntos
Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/patologia , Fármacos Neuroprotetores/administração & dosagem , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Infecção por Zika virus/complicações , Infecção por Zika virus/patologia , Zika virus/crescimento & desenvolvimento , Animais , Modelos Animais de Doenças , Camundongos , Resultado do Tratamento
13.
J Immunol ; 198(10): 4096-4106, 2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28424241

RESUMO

The indigenous intestinal microbiota is frequently considered an additional major organ of the human body and exerts profound immunomodulating activities. Germ-free (GF) mice display a significantly different inflammatory responsiveness pattern compared with conventional (CV) mice, and this was dubbed a "hyporesponsive phenotype." Taking into account that the deposition of immune complexes is a major event in acute inflammation and that GF mice have a distinct Ig repertoire and B cell activity, we aimed to evaluate whether this altered Ig repertoire interferes with the inflammatory responsiveness of GF mice. We found that serum transfer from CV naive mice was capable of reversing the inflammatory hyporesponsiveness of GF mice in sterile inflammatory injury induced by intestinal ischemia and reperfusion, as well as in a model of lung infection by Klebsiella pneumoniae Transferring serum from Ig-deficient mice to GF animals did not alter their response to inflammatory insult; however, injecting purified Abs from CV animals restored inflammatory responsiveness in GF mice, suggesting that natural Abs present in serum were responsible for altering GF responsiveness. Mechanistically, injection of serum and Ig from CV mice into GF animals restored IgG deposition, leukocyte influx, NF-κB activation, and proinflammatory gene expression in inflamed tissues and concomitantly downregulated annexin-1 and IL-10 production. Thus, our data show that microbiota-induced natural Abs are pivotal for host inflammatory responsiveness to sterile and infectious insults.


Assuntos
Anticorpos/imunologia , Microbioma Gastrointestinal/imunologia , Vida Livre de Germes , Inflamação/imunologia , Intestinos/imunologia , Animais , Anexinas/imunologia , Anticorpos/administração & dosagem , Linfócitos B/imunologia , Regulação da Expressão Gênica , Humanos , Interleucina-10/imunologia , Intestinos/microbiologia , Intestinos/patologia , Isquemia , Infecções por Klebsiella/imunologia , Infecções por Klebsiella/microbiologia , Klebsiella pneumoniae/imunologia , Pulmão/imunologia , Pulmão/microbiologia , Camundongos , NF-kappa B/genética
14.
J Neuroinflammation ; 14(1): 61, 2017 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-28330482

RESUMO

BACKGROUND: Flaviviruses are a genre of closely related viral pathogens which emerged in the last decades in Brazil and in the world. Saint (St.) Louis encephalitis virus (SLEV) is a neglected flavivirus that can cause a severe neurological disease that may lead to death or sequelae. St. Louis encephalitis pathogenesis is poorly understood, which hinders the development of specific treatment or vaccine. METHODS: To address this problem, we developed a model of SLEV infection in mice to study mechanisms involved in the pathogenesis of severe disease. The model consists in the intracranial inoculation of the SLEV strain BeH 355964, a strain isolated from a symptomatic human patient in Brazil, in adult immunocompetent mice. RESULTS: Inoculated mice presented SLEV replication in the brain, accompanied by tissue damage, disease signs, and mortality approximately 7 days post infection. Infection was characterized by the production of proinflammatory cytokines and interferons and by leukocyte recruitment to the brain, composed mainly by neutrophils and lymphocytes. In vitro experiments indicated that SLEV is able to replicate in both neurons and glia and caused neuronal death and cytokine production, respectively. CONCLUSIONS: Altogether, intracranial SLEV infection leads to meningoencephalitis in mice, recapitulating several aspects of St. Louis encephalitis in humans. Our study indicates that the central nervous system (CNS) inflammation is a major component of SLEV-induced disease. This model may be useful to identify mechanisms of disease pathogenesis or resistance to SLEV infection.


Assuntos
Citocinas/metabolismo , Modelos Animais de Doenças , Vírus da Encefalite de St. Louis/fisiologia , Encefalite de St. Louis/patologia , Análise de Variância , Animais , Linhagem Celular Transformada , Encefalite de St. Louis/virologia , Peroxidase de Eosinófilo/metabolismo , Hexosaminidases/metabolismo , Leucócitos/metabolismo , Leucócitos/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Peroxidase/metabolismo , Fatores de Tempo , Carga Viral
15.
Artigo em Inglês | MEDLINE | ID: mdl-27867403

RESUMO

Stryphnodendron species, popularly named "barbatimão," are traditionally used in Brazil as anti-inflammatory agents. This study aimed to investigate the effect of barbatimão and 11 other species on the production of tumor necrosis factor-alpha (TNF-α) in lipopolysaccharide- (LPS-) stimulated THP-1 cells, as well as their anti-arthritis activity. The extracts of Stryphnodendron adstringens, Stryphnodendron obovatum, Campomanesia lineatifolia, and Terminalia glabrescens promoted a concentration-dependent inhibition of TNF-α. Mice injected with LPS in the knee joint were treated per os with fractions from the selected extracts. Both the organic (SAO) and the aqueous (SAA) fractions of S. adstringens promoted a dose-dependent reduction of leukocyte migration and neutrophil accumulation into the joint, but none of them reduced CXCL1 concentration in the periarticular tissue. In contrast, treatment with C. lineatifolia and T. glabrescens fractions did not ameliorate the inflammatory parameters. Analyses of SAO by Ultra Performance Liquid Chromatography (UPLC) coupled to electrospray ionization mass spectrometry (ESI-MS) led to the identification of gallic acid along with 11 prodelphinidins, characterized as monomers and dimers of the B-type. Our findings contribute to some extent to corroborating the traditional use of S. adstringens as an anti-inflammatory agent. This activity is probably related to a decrease of leukocyte migration into the inflammatory site. Polyphenols like gallic acid and prodelphinidins, identified in the active fraction, may contribute to the observed activity.

16.
Infect Immun ; 84(10): 3071-82, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27481250

RESUMO

The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor involved in controlling several aspects of immune responses, including the activation and differentiation of specific T cell subsets and antigen-presenting cells, thought to be relevant in the context of experimental Trypanosoma cruzi infection. The relevance of AhR for the outcome of T. cruzi infection is not known and was investigated here. We infected wild-type (WT) mice and AhR knockout (AhR KO) mice with T. cruzi (Y strain) and determined levels of parasitemia, myocardial inflammation and fibrosis, expression of AhR/cytokines/suppressor of cytokine signaling (SOCS) (spleen/heart), and production of nitric oxide (NO), reactive oxygen species (ROS), and peroxynitrite (ONOO(-)) (spleen). AhR expression was increased in the heart of infected WT mice. Infected AhR KO mice displayed significantly reduced parasitemia, inflammation, and fibrosis of the myocardium. This was associated with an anticipated increased immune response characterized by increased levels of inflammatory cytokines and reduced expression of SOCS2 and SOCS3 in the heart. In vitro, AhR deficiency caused impairment in parasite replication and decreased levels of ROS production. In conclusion, AhR influences the development of murine Chagas disease by modulating ROS production and regulating the expression of key physiological regulators of inflammation, SOCS1 to -3, associated with the production of cytokines during experimental T. cruzi infection.


Assuntos
Doença de Chagas/fisiopatologia , Citocinas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores de Hidrocarboneto Arílico/fisiologia , Trypanosoma cruzi/fisiologia , Animais , Cardiomiopatia Chagásica/metabolismo , Cardiomiopatia Chagásica/patologia , Doença de Chagas/metabolismo , Doença de Chagas/patologia , Modelos Animais de Doenças , Camundongos , Camundongos Knockout , Miocardite/metabolismo , Miocardite/patologia , Óxido Nítrico/metabolismo , Ácido Peroxinitroso/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo , Baço/metabolismo , Proteínas Supressoras da Sinalização de Citocina/metabolismo
17.
Environ Entomol ; 45(5): 1161-1169, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27550163

RESUMO

Impacts of habitat loss and fragmentation on specialist herbivores have been rarely addressed. Here we examine the structure of plant and galling insect assemblages in a fragmented landscape of the Atlantic forest to verify a potential impoverishment of these assemblages mediated by edge effects. Saplings and galling insects were recorded once within a 0.1-ha area at habitat level, covering forest interior stands, forest edges, and small fragments. A total of 1,769 saplings from 219 tree species were recorded across all three habitats, with differences in terms of sapling abundance and species richness. Additionally, edge-affected habitats exhibited reduced richness of both host-plant and galling insects at plot and habitat spatial scale. Attack levels also differed among forest types at habitat spatial scale (21.1% of attacked stems in forest interior, 12.4% in small fragments but only 8.5% in forest edges). Plot ordination resulted in three clearly segregated clusters: one formed by forest interior, one by small fragments, and another formed by edge plots. Finally, the indicator species analysis identified seven and one indicator plant species in forest interior and edge-affected habitats, respectively. Consequently, edge effects lead to formation of distinct taxonomic groups and also an impoverished assemblage of plants and galling insects at multiple spatial scales. The results of the present study indicate that fragmentation-related changes in plant assemblages can have a cascade effects on specialist herbivores. Accordingly, hyperfragmented landscapes may not be able to retain an expressive portion of tropical biodiversity.


Assuntos
Biodiversidade , Florestas , Herbivoria , Insetos/fisiologia , Árvores/fisiologia , Animais , Brasil
18.
Am J Hypertens ; 29(10): 1179-85, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27179254

RESUMO

BACKGROUND: Excessive inflammation is involved in preeclampsia (PE) pathogenesis. Lipoxin A4 (LXA4) is an eicosanoid that counter-regulates inflammation. The main objective of this study was to determine LXA4 plasma levels in PE women. The correlations among LXA4 levels, ultrasensitive C-reactive protein (us-CRP) levels, and clinical/laboratory parameters of the studied participants were also investigated. METHODS: LXA4 plasma levels were determined by ELISA in 23 nonpregnant, 26 normotensive pregnant, and 27 PE women (early PE (N = 10) and late PE (N = 17)), according to gestational age (GA) at clinical symptoms onset). The clinical/laboratory parameters included in Spearman's correlation analysis were: systolic and diastolic blood pressure (SBP and DBP, respectively), lactate dehydrogenase (LDH) activity, platelet count, proteinuria, and white blood cell count (WBC). RESULTS: LXA4 levels were higher in PE women than in nonpregnant and normotensive pregnant women, and similar between nonpregnant and normotensive pregnant women. LXA4 plasma levels were higher in early PE vs. normotensive pregnancy (GA < 34 weeks) and in late PE vs. normotensive pregnancy (GA ≥ 34 weeks). No significant differences were detected between early and late PE. LXA4 levels were positively correlated with us-CRP levels, SBP, DBP, and WBC. No significant correlation was detected between LXA4 levels and the other laboratory parameters. CONCLUSIONS: Chronic inflammation in PE, in spite of increased levels of LXA4, points to a possible failure in this regulatory pathway. Further studies are necessary to clarify this issue and to evaluate the role of LXA4 and other proresolving mediators of inflammation in the pathogenesis of PE.


Assuntos
Lipoxinas/sangue , Pré-Eclâmpsia/sangue , Adulto , Proteína C-Reativa/metabolismo , Estudos de Casos e Controles , Feminino , Humanos , Pré-Eclâmpsia/imunologia , Gravidez , Adulto Jovem
19.
Int J Med Microbiol ; 306(4): 187-95, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27083265

RESUMO

The inflammatory response plays a crucial role in infectious diseases, and the intestinal microbiota is linked to maturation of the immune system. However, the association between microbiota and the response against fungal infections has not been elucidated. Our aim was to evaluate the influence of microbiota on Cryptococcus gattii infection. Germ-free (GF), conventional (CV), conventionalized (CVN-mice that received feces from conventional animals), and LPS-stimulated mice were infected with C. gattii. GF mice were more susceptible to infection, showing lower survival, higher fungal burden in the lungs and brain, increased behavioral changes, reduced levels of IFN-γ, IL-1ß and IL-17, and lower NFκBp65 phosphorylation compared to CV mice. Low expression of inflammatory cytokines was associated with smaller yeast cells and polysaccharide capsules (the main virulence factor of C. gattii) in the lungs, and less tissue damage. Furthermore, macrophages from GF mice showed reduced ability to engulf, produce ROS, and kill C. gattii. Restoration of microbiota (CVN mice) or LPS administration made GF mice more responsive to infection, which was associated with increased survival and higher levels of inflammatory mediators. This study is the first to demonstrate the influence of microbiota in the host response against C. gattii.


Assuntos
Criptococose/imunologia , Criptococose/patologia , Cryptococcus gattii/imunologia , Suscetibilidade a Doenças , Microbioma Gastrointestinal/imunologia , Inflamação/patologia , Animais , Proteínas Reguladoras de Apoptose , Encéfalo/microbiologia , Encéfalo/patologia , Contagem de Colônia Microbiana , Citocinas/metabolismo , Modelos Animais de Doenças , Vida Livre de Germes , Pulmão/microbiologia , Pulmão/patologia , Macrófagos/imunologia , Camundongos , Fagocitose , Receptores Imunológicos , Receptores Depuradores , Análise de Sobrevida , Proteína da Síndrome de Wiskott-Aldrich
20.
Pharmacol Res ; 112: 68-83, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27107789

RESUMO

Immune responses are fundamental for protecting against most infectious agents. However, there is now much evidence to suggest that the pathogenesis and tissue damage after infection are not usually related to the direct action of the replication of microorganisms, but instead to altered immune responses triggered after the contact with the pathogen. This review article discusses several mechanisms necessary for the host to protect against microbial infection and focuses in aspects that cause altered inflammation and drive immunopathology. These basic findings can ultimately reveal pathways amenable to host-directed therapy in adjunct to antimicrobial therapy for future improved control measures for many infectious diseases. Therefore, modulating the effects of inflammatory pathways may represent a new therapy during infection outcome and disease.


Assuntos
Anti-Infecciosos/farmacologia , Anti-Infecciosos/uso terapêutico , Interações Hospedeiro-Patógeno , Infecções/tratamento farmacológico , Infecções/imunologia , Animais , Anti-Infecciosos/imunologia , Bactérias/efeitos dos fármacos , Bactérias/metabolismo , Infecções Bacterianas/tratamento farmacológico , Infecções Bacterianas/imunologia , Descoberta de Drogas , Humanos , Inflamação/tratamento farmacológico , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/microbiologia , Camundongos , Terapia de Alvo Molecular , Micoses/tratamento farmacológico , Micoses/imunologia , Viroses/tratamento farmacológico , Viroses/imunologia , Vírus/efeitos dos fármacos , Vírus/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...