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

País/Región como asunto
Intervalo de año de publicación
1.
Nat Mater ; 17(11): 971-977, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30349030

RESUMEN

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.


Asunto(s)
Antivirales/uso terapéutico , Encéfalo/metabolismo , Péptidos/uso terapéutico , Infección por el Virus Zika/tratamiento farmacológico , Animales , Antivirales/farmacocinética , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Péptidos/farmacocinética
2.
J Immunol ; 198(10): 4096-4106, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28424241

RESUMEN

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.


Asunto(s)
Anticuerpos/inmunología , Microbioma Gastrointestinal/inmunología , Vida Libre de Gérmenes , Inflamación/inmunología , Intestinos/inmunología , Animales , Anexinas/inmunología , Anticuerpos/administración & dosificación , Linfocitos B/inmunología , Regulación de la Expresión Génica , Humanos , Interleucina-10/inmunología , Intestinos/microbiología , Intestinos/patología , Isquemia , Infecciones por Klebsiella/inmunología , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/inmunología , Pulmón/inmunología , Pulmón/microbiología , Ratones , FN-kappa B/genética
3.
Immunology ; 155(4): 477-490, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30098206

RESUMEN

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.


Asunto(s)
Virus del Dengue/inmunología , Interleucina-33/farmacología , Receptores de Interleucina-8B/antagonistas & inhibidores , Proteínas Recombinantes/farmacología , Animales , Dengue/inmunología , Dengue/virología , Progresión de la Enfermedad , Proteína 1 Similar al Receptor de Interleucina-1/deficiencia , Proteína 1 Similar al Receptor de Interleucina-1/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Sulfonamidas/farmacología
4.
Artículo en Inglés | MEDLINE | ID: mdl-29463546

RESUMEN

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.


Asunto(s)
Antibacterianos/uso terapéutico , Atorvastatina/uso terapéutico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Imipenem/uso terapéutico , Infecciones por Klebsiella/tratamiento farmacológico , Klebsiella pneumoniae/efectos de los fármacos , Neumonía Bacteriana/tratamiento farmacológico , Animales , Carga Bacteriana/efectos de los fármacos , Quimiocinas/análisis , Infecciones Comunitarias Adquiridas/tratamiento farmacológico , Infecciones Comunitarias Adquiridas/microbiología , Farmacorresistencia Bacteriana Múltiple , Quimioterapia Combinada , Femenino , Inflamación/tratamiento farmacológico , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Neutrófilos/inmunología , Fagocitosis/efectos de los fármacos , Fagocitosis/inmunología , Neumonía Bacteriana/microbiología
5.
Eur J Immunol ; 46(1): 204-11, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26449770

RESUMEN

Gout manifests as recurrent episodes of acute joint inflammation and pain due to the deposition of monosodium urate (MSU) crystals within the affected tissue in a process dependent on NLRP3 inflammasome activation. The synthesis, activation, and release of IL-1ß are crucial for MSU-induced inflammation. The current study evaluated the mechanism by which TNF-α contributed to MSU-induced inflammation. Male C57BL/6J or transgenic mice were used in this study and inflammation was induced by the injection of MSU crystals into the joint. TNF-α was markedly increased in the joint after the injection of MSU. There was inhibition in the infiltration of neutrophils, production of CXCL1 and IL-1ß, and decreased hypernociception in mice deficient for TNF-α or its receptors. Pharmacological blockade of TNF-α with Etanercept or pentoxyfylline produced similar results. Mechanistically, TNF-α blockade resulted in lower amounts of IL-1ß protein and pro-IL-1ß mRNA transcripts in joints. Gene-modified mice that express only transmembrane TNF-α had an inflammatory response similar to that of WT mice and blockade of soluble TNF-α (XPro™1595) did not decrease MSU-induced inflammation. In conclusion, TNF-α drives expression of pro-IL-1ß mRNA and IL-1ß protein in experimental gout and that its transmembrane form is sufficient to trigger MSU-induced inflammation in mice.


Asunto(s)
Gota/inmunología , Hiperalgesia/etiología , Inflamación/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Modelos Animales de Enfermedad , Gota/complicaciones , Gota/metabolismo , Inflamación/metabolismo , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Articulación de la Rodilla , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Estimulación Física , Reacción en Cadena en Tiempo Real de la Polimerasa , Ácido Úrico/efectos adversos , Ácido Úrico/inmunología
6.
J Neuroinflammation ; 14(1): 61, 2017 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-28330482

RESUMEN

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.


Asunto(s)
Citocinas/metabolismo , Modelos Animales de Enfermedad , Virus de la Encefalitis de San Luis/fisiología , Encefalitis de San Luis/patología , Análisis de Varianza , Animales , Línea Celular Transformada , Encefalitis de San Luis/virología , Peroxidasa del Eosinófilo/metabolismo , Hexosaminidasas/metabolismo , Leucocitos/metabolismo , Leucocitos/virología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Peroxidasa/metabolismo , Factores de Tiempo , Carga Viral
7.
Infect Immun ; 84(10): 3071-82, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27481250

RESUMEN

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.


Asunto(s)
Enfermedad de Chagas/fisiopatología , Citocinas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores de Hidrocarburo de Aril/fisiología , Trypanosoma cruzi/fisiología , Animales , Cardiomiopatía Chagásica/metabolismo , Cardiomiopatía Chagásica/patología , Enfermedad de Chagas/metabolismo , Enfermedad de Chagas/patología , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados , Miocarditis/metabolismo , Miocarditis/patología , Óxido Nítrico/metabolismo , Ácido Peroxinitroso/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Bazo/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo
8.
Int J Med Microbiol ; 306(4): 187-95, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27083265

RESUMEN

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.


Asunto(s)
Criptococosis/inmunología , Criptococosis/patología , Cryptococcus gattii/inmunología , Susceptibilidad a Enfermedades , Microbioma Gastrointestinal/inmunología , Inflamación/patología , Animales , Proteínas Reguladoras de la Apoptosis , Encéfalo/microbiología , Encéfalo/patología , Recuento de Colonia Microbiana , Citocinas/metabolismo , Modelos Animales de Enfermedad , Vida Libre de Gérmenes , Pulmón/microbiología , Pulmón/patología , Macrófagos/inmunología , Ratones , Fagocitosis , Receptores Inmunológicos , Receptores Depuradores , Análisis de Supervivencia , Proteína del Síndrome de Wiskott-Aldrich
9.
Pharmacol Res ; 112: 68-83, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27107789

RESUMEN

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.


Asunto(s)
Antiinfecciosos/farmacología , Antiinfecciosos/uso terapéutico , Interacciones Huésped-Patógeno , Infecciones/tratamiento farmacológico , Infecciones/inmunología , Animales , Antiinfecciosos/inmunología , Bacterias/efectos de los fármacos , Bacterias/metabolismo , Infecciones Bacterianas/tratamiento farmacológico , Infecciones Bacterianas/inmunología , Descubrimiento de Drogas , Humanos , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/microbiología , Ratones , Terapia Molecular Dirigida , Micosis/tratamiento farmacológico , Micosis/inmunología , Virosis/tratamiento farmacológico , Virosis/inmunología , Virus/efectos de los fármacos , Virus/metabolismo
10.
Immunol Rev ; 245(1): 250-64, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22168425

RESUMEN

Mammals are subject to colonization by an astronomical number of mutualistic and commensal microorganisms on their environmental exposed surfaces. These mutualistic species build up a complex community, called the indigenous microbiota, which aid their hosts in several physiological activities. In this review, we show that the transition between a non-colonized and a colonized state is associated with modification on the pattern of host inflammatory and behavioral responsiveness. There is a shift from innate anti-inflammatory cytokine production to efficient release of proinflammatory mediators and rapid mobilization of leukocytes upon infection or other stimuli. In addition, host responses to hypernociceptive and stressful stimuli are modulated by indigenous microbiota, partly due to the altered pattern of innate and acquired immune responsiveness of the non-colonized host. These altered responses ultimately lead to significant alteration in host behavior to environmental threats. Therefore, host colonization by indigenous microbiota modifies the way the host perceives and reacts to environmental stimuli, improving resilience of the entire host-microorganism consortium to environmental stresses.


Asunto(s)
Infecciones Bacterianas/inmunología , Infecciones Bacterianas/psicología , Conducta , Inmunidad Innata , Nociceptores/inmunología , Estrés Fisiológico/inmunología , Adaptación Biológica , Animales , Infecciones Bacterianas/microbiología , Interacciones Huésped-Patógeno , Humanos , Inflamación/inmunología
11.
Immunology ; 145(4): 583-96, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25939314

RESUMEN

Dengue is a mosquito-borne disease that affects millions of people worldwide yearly. Currently, there is no vaccine or specific treatment available. Further investigation on dengue pathogenesis is required to better understand the disease and to identify potential therapeutic targets. The chemokine system has been implicated in dengue pathogenesis, although the specific role of chemokines and their receptors remains elusive. Here we describe the role of the CC-chemokine receptor CCR5 in Dengue virus (DENV-2) infection. In vitro experiments showed that CCR5 is a host factor required for DENV-2 replication in human and mouse macrophages. DENV-2 infection induces the expression of CCR5 ligands. Incubation with an antagonist prevents CCR5 activation and reduces DENV-2 positive-stranded (+) RNA inside macrophages. Using an immunocompetent mouse model of DENV-2 infection we found that CCR5(-/-) mice were resistant to lethal infection, presenting at least 100-fold reduction of viral load in target organs and significant reduction in disease severity. This phenotype was reproduced in wild-type mice treated with CCR5-blocking compounds. Therefore, CCR5 is a host factor required for DENV-2 replication and disease development. Targeting CCR5 might represent a therapeutic strategy for dengue fever. These data bring new insights on the association between viral infections and the chemokine receptor CCR5.


Asunto(s)
Virus del Dengue/fisiología , Dengue/inmunología , Macrófagos/inmunología , Receptores CCR5/inmunología , Replicación Viral/inmunología , Animales , Secuencia de Bases , Dengue/tratamiento farmacológico , Dengue/genética , Humanos , Macrófagos/patología , Macrófagos/virología , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Receptores CCR5/genética , Replicación Viral/efectos de los fármacos , Replicación Viral/genética
12.
Antimicrob Agents Chemother ; 59(7): 4003-11, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25896704

RESUMEN

Paracoccidioidomycosis (PCM) is a public health concern in Latin America and South America that when not correctly treated can lead to patient death. In this study, the influence of melanin produced by Paracoccidioides spp. on the effects of treatment with antimicrobial photodynamic inhibition (aPI) and antifungal drugs was evaluated. aPI was performed using toluidine blue (TBO) as a photosensitizer and a 630-nm light-emitting diode (LED) light. The antifungals tested were itraconazole and amphotericin B. We evaluated the effects of each approach, aPI or antifungals, against nonmelanized and melanized yeast cells by performing susceptibility tests and by quantifying oxidative and nitrosative bursts during the experiments. aPI reduced nonmelanized cells by 3.0 log units and melanized cells by 1.3 log units. The results showed that melanization protects the fungal cell, probably by acting as a scavenger of nitric oxide and reactive oxygen species, but not of peroxynitrite. Melanin also increased the MICs of itraconazole and amphotericin B, and the drugs were fungicidal for nonmelanized and fungistatic for melanized yeast cells. Our study shows that melanin production by Paracoccidioides yeast cells serves a protective function during aPI and treatment with itraconazole and amphotericin B. The results suggest that melanin binds to the drugs, changing their antifungal activities, and also acts as a scavenger of reactive oxygen species and nitric oxide, but not of peroxynitrite, indicating that peroxynitrite is the main radical that is responsible for fungal death after aPI.


Asunto(s)
Antifúngicos/farmacología , Melaninas/farmacología , Paracoccidioides/efectos de los fármacos , Fotoquimioterapia , Anfotericina B/química , Anfotericina B/farmacología , Antifúngicos/química , Farmacorresistencia Fúngica/efectos de los fármacos , Depuradores de Radicales Libres/farmacología , Itraconazol/química , Itraconazol/farmacología , Lacasa/metabolismo , Levodopa/farmacología , Melaninas/química , Pruebas de Sensibilidad Microbiana , Óxido Nítrico/metabolismo , Ácido Peroxinitroso/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Estallido Respiratorio/efectos de los fármacos
13.
Am J Pathol ; 184(7): 2023-34, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24952429

RESUMEN

Irinotecan is a useful chemotherapeutic for the treatment of various cancers. Irinotecan treatment is associated with mucositis, which clearly limits the use of the drug. Mechanisms that account for mucositis are only partially known. This study assessed mechanisms and the role of inflammasome activation in irinotecan-induced mucositis. Mucositis in mice was induced by irinotecan injection in C57BL/6 wild-type, gp91phox(-/-), il-18(-/-), casp-1(-/-), and asc(-/-) mice once a day for 4 consecutive days. In some experiments, mice received apocynin to inhibit NADPH oxidase (NOX), IL-1 receptor antagonist, or IL-18 binding protein to prevent activation of IL-1 and IL-18 receptors, respectively. Mice were euthanized 7 days after the beginning of irinotecan treatment, and small intestines were collected for analysis. Irinotecan treatment resulted in increased IL-1ß and IL-18 production in ileum and NOX-2-dependent oxidative stress. gp91phox(-/-) and apocynin-treated mice had diminished oxidative stress and less severe mucositis. Furthermore, treatment with apocynin decreased caspase-1 activation and IL-1ß and IL-18 production in the ileum. asc(-/-) and casp-1(-/-) mice also had less intestinal injury and decreased IL-1ß and IL-18 production. Finally, both the absence of IL-18 and IL-1ß resulted in reduced inflammatory response and attenuated intestinal injury. NOX-2-derived oxidative stress mediates inflammasome activation and inflammasome-dependent production of IL-1ß and IL-18, which mediate tissue injury during irinotecan-induced mucositis in mice.


Asunto(s)
Inflamasomas/metabolismo , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Mucositis/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Camptotecina/efectos adversos , Camptotecina/análogos & derivados , Caspasa 1/metabolismo , Íleon/metabolismo , Íleon/patología , Irinotecán , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Mucositis/inducido químicamente , NADPH Oxidasa 2 , NADPH Oxidasas/metabolismo , Estrés Oxidativo
14.
Phytother Res ; 29(12): 1887-93, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26344935

RESUMEN

The leaves of Hancornia speciosa Gomes (Apocynaceae), a medicinal species found in the Brazilian cerrado biome, are traditionally used to treat wounds and inflammatory disorders. The goal of the present study was to investigate the in vitro wound healing properties of ethanolic extract of H. speciosa leaves and its isolated compounds, using the scratch assay, and to evaluate their effects on the release of the pro-inflammatory cytokine tumor necrosis factor (TNF-α) by lipopolysaccharide (LPS)-stimulated human acute monocytic (THP-1) cells. H. speciosa ethanolic extract significantly increased (42.8% ± 5.4 at 25 µg/mL) cell migration and proliferation of fibroblasts compared with control cells, as well as the isolated compounds bornesitol (80.8% ± 5.1) and quinic acid (69.1% ± 6.2), both assayed at 50 µM. TNF-α release by LPS-stimulated THP-1 cells was significantly reduced by the ethanolic extract (62.9% ± 8.2, i.e. 1791.1 ± 394.7 pg/mL) at 10 µg/mL, bornesitol (48.9% ± 0.9, i.e. 2461.6 ± 43.1 pg/mL) at 50 µM, and quinic acid (90.2% ± 3.4, i.e. 473.5 ± 164.4 pg/mL) and rutin (82.4% ± 5.6, i.e. 847.0 ± 271.8 pg/mL) at 10 µM. These results provided evidences to support the traditional use of H. speciosa leaves to treat wounds and inflammatory disorders.


Asunto(s)
Apocynaceae/química , Fibroblastos/efectos de los fármacos , Extractos Vegetales/farmacología , Cicatrización de Heridas/efectos de los fármacos , Brasil , Línea Celular , Ciclitoles/aislamiento & purificación , Ciclitoles/farmacología , Humanos , Lipopolisacáridos , Hojas de la Planta/química , Ácido Quínico/aislamiento & purificación , Ácido Quínico/farmacología , Rutina/aislamiento & purificación , Rutina/farmacología , Factor de Necrosis Tumoral alfa/metabolismo
15.
Infect Immun ; 82(8): 3127-40, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24818665

RESUMEN

Infection with Plasmodium falciparum may result in severe disease affecting various organs, including liver, spleen, and brain, resulting in high morbidity and mortality. Plasmodium berghei Anka infection of mice recapitulates many features of severe human malaria. The aryl hydrocarbon receptor (AhR) is an intracellular receptor activated by ligands important in the modulation of the inflammatory response. We found that AhR-knockout (KO) mice infected with P. berghei Anka displayed increased parasitemia, earlier mortality, enhanced leukocyte-endothelial cell interactions in the brain microvasculature, and increased inflammation in brain (interleukin-17 [IL-17] and IL-6) and liver (gamma interferon [IFN-γ] and tumor necrosis factor alpha [TNF-α]) compared to infected wild-type (WT) mice. Infected AhR-KO mice also displayed a reduction in cytokines required for host resistance, including TNF-α, IL-1ß, and IFN-γ, in the brain and spleen. Infection of AhR-KO mice resulted in an increase in T regulatory cells and transforming growth factor ß, IL-6, and IL-17 in the brain. AhR modulated the basal expression of SOCS3 in spleen and brain, and P. berghei Anka infection resulted in enhanced expression of SOCS3 in brain, which was absent in infected AhR-KO mice. These data suggest that AhR-mediated control of SOCS3 expression is probably involved in the phenotype seen in infected AhR-KO mice. This is, to our knowledge, the first demonstration of a role for AhR in the pathogenesis of malaria.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/inmunología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Malaria/inmunología , Malaria/patología , Plasmodium berghei/inmunología , Receptores de Hidrocarburo de Aril/inmunología , Receptores de Hidrocarburo de Aril/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/inmunología , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Encéfalo/inmunología , Encéfalo/patología , Citocinas/inmunología , Citocinas/metabolismo , Eliminación de Gen , Humanos , Hígado/inmunología , Hígado/patología , Malaria/parasitología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Hidrocarburo de Aril/genética , Bazo/inmunología , Proteína 3 Supresora de la Señalización de Citocinas
16.
Eur J Immunol ; 43(6): 1529-44, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23505056

RESUMEN

Dengue virus (DENV), a mosquito-borne flavivirus, is a public health problem in many tropical countries. IL-22 and IL-17A are key cytokines in several infectious and inflammatory diseases. We have assessed the contribution of IL-22 and IL-17A in the pathogenesis of experimental dengue infection using a mouse-adapted DENV serotype 2 strain (P23085) that causes a disease that resembles severe dengue in humans. We show that IL-22 and IL-17A are produced upon DENV-2 infection in immune-competent mice. Infected IL-22(-/-) mice had increased lethality, neutrophil accumulation and pro-inflammatory cytokines in tissues, notably IL-17A. Viral load was increased in spleen and liver of infected IL-22(-/-) mice. There was also more severe liver injury, as seen by increased transaminases levels and tissue histopathology. γδ T cells and NK cells are sources of IL-17A and IL-22, respectively, in liver and spleen. We also show that DENV-infected HepG2 cells treated with rhIL-22 had reduced cell death and decreased IL-6 production. IL-17RA(-/-) mice were protected upon infection and IL-17A-neutralizing-Ab-treatment partially reversed the phenotype observed in IL-22(-/-) -infected mice. We suggest that disrupting the balance between IL-22 and IL-17A levels may represent an important strategy to reduce inflammation and tissue injury associated with severe dengue infection.


Asunto(s)
Virus del Dengue/inmunología , Dengue/inmunología , Mediadores de Inflamación/metabolismo , Interleucina-17/metabolismo , Interleucinas/metabolismo , Hígado/metabolismo , Neutrófilos/inmunología , Animales , Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Células Hep G2 , Humanos , Inflamación/genética , Interleucina-17/genética , Interleucina-6/genética , Interleucina-6/metabolismo , Interleucinas/genética , Interleucinas/inmunología , Hígado/inmunología , Hígado/patología , Hígado/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/virología , Receptores de Interleucina-17/genética , Carga Viral/genética , Interleucina-22
17.
Am J Pathol ; 182(6): 1950-61, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23567637

RESUMEN

Dengue disease is a mosquito-borne viral disease of expanding geographical range and incidence. Infection by one of the four serotypes of dengue virus induces a spectrum of disease manifestations, ranging from asymptomatic to life-threatening Dengue hemorrhagic fever/dengue shock syndrome. Many efforts have been made to elucidate several aspects of dengue virus-induced disease, but the pathogenesis of disease is complex and remains unclear. Understanding the mechanisms involved in the early stages of infection is crucial to determine and develop safe therapeutics to prevent the severe outcomes of disease without interfering with control of infection. In this review, we discuss the dual role of the innate and inflammatory pathways activated during dengue disease in mediating both protection and exacerbation of disease. We show that some mediators involved in each of these responses differ substantially, suggesting that interfering in disease-associated immune pathways may represent a potential therapeutic opportunity for the treatment of severe dengue.


Asunto(s)
Dengue/inmunología , Inflamación/virología , Animales , Citocinas/inmunología , Dengue/complicaciones , Dengue/fisiopatología , Dengue/prevención & control , Modelos Animales de Enfermedad , Humanos , Inmunidad Innata , Ratones
18.
Med Microbiol Immunol ; 203(4): 231-50, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24723052

RESUMEN

Dengue is a mosquito-borne disease caused by one of four serotypes of Dengue virus (DENV-1-4). Epidemiologic and observational studies demonstrate that the majority of severe dengue cases, dengue hemorrhagic fever and dengue shock syndrome (DHF/DSS), occurs predominantly in either individuals with cross-reactive immunity following a secondary heterologous infection or in infants with primary DENV infections born from dengue-immune mothers, suggesting that B-cell-mediated and antibody responses impact on disease evolution. We demonstrate here that B cells play a pivotal role in host responses against primary DENV infection in mice. After infection, µMT(-/-) mice showed increased viral loads followed by severe disease manifestation characterized by intense thrombocytopenia, hemoconcentration, cytokine production and massive liver damage that culminated in death. In addition, we show that poly and monoclonal anti-DENV-specific antibodies can sufficiently increase viral replication through a suppression of early innate antiviral responses and enhance disease manifestation, so that a mostly non-lethal illness becomes a fatal disease resembling human DHF/DSS. Finally, treatment with intravenous immunoglobulin containing anti-DENV antibodies confirmed the potential enhancing capacity of subneutralizing antibodies to mediate virus infection and replication and induce severe disease manifestation of DENV-infected mice. Thus, our results show that humoral responses unleashed during DENV infections can exert protective or pathological outcomes and provide insight into the pathogenesis of this important human pathogen.


Asunto(s)
Acrecentamiento Dependiente de Anticuerpo , Virus del Dengue/inmunología , Dengue/inmunología , Dengue/patología , Inmunidad Innata , Animales , Linfocitos B/inmunología , Citocinas/sangre , Muerte , Hígado/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Trombocitopenia , Carga Viral
19.
J Immunol ; 188(3): 1411-20, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22210917

RESUMEN

Mammals are colonized by an astronomical number of commensal microorganisms on their environmental exposed surfaces. These symbiotic species build up a complex community that aids their hosts in several physiological activities. We have shown that lack of intestinal microbiota is accompanied by a state of active IL-10-mediated inflammatory hyporesponsiveness. The present study investigated whether the germfree state and its hyporesponsive phenotype alter host resistance to an infectious bacterial insult. Experiments performed in germfree mice infected with Klebsiella pneumoniae showed that these animals are drastically susceptible to bacterial infection in an IL-10-dependent manner. In germfree mice, IL-10 restrains proinflammatory mediator production and neutrophil recruitment and favors pathogen growth and dissemination. Germfree mice were resistant to LPS treatment. However, priming of these animals with several TLR agonists recovered their inflammatory responsiveness to sterile injury. LPS pretreatment also rendered germfree mice resistant to pulmonary K. pneumoniae infection, abrogated IL-10 production, and restored TNF-α and CXCL1 production and neutrophil mobilization into lungs of infected germfree mice. This effective inflammatory response mounted by LPS-treated germfree mice resulted in bacterial clearance and enhanced survival upon infection. Therefore, host colonization by indigenous microbiota alters the way the host reacts to environmental infectious stimuli, probably through activation of TLR-dependent pathways. Symbiotic gut colonization enables proper inflammatory response to harmful insults to the host, and increases resilience of the entire mammal-microbiota consortium to environmental pressures.


Asunto(s)
Inflamación/etiología , Infecciones por Klebsiella/inmunología , Receptores Toll-Like/metabolismo , Animales , Vida Libre de Gérmenes , Interacciones Huésped-Patógeno/inmunología , Interleucina-10/inmunología , Intestinos/inmunología , Intestinos/microbiología , Klebsiella pneumoniae , Metagenoma/inmunología , Ratones , Receptores Toll-Like/fisiología
20.
Planta Med ; 80(8-9): 630-6, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24871207

RESUMEN

Azadirachta indica (Meliaceae) extracts have been reported to exhibit anti-inflammatory and antinociceptive properties. However, the activities of azadirachtin, a limonoid and the major bioactive compound found in the extracts, have been poorly investigated in animal models. In the present study, we investigated the effects induced by azadirachtin in experimental models of pain and inflammation in mice. Carrageenan-induced paw edema and fibrovascular tissue growth induced by subcutaneous cotton pellet implantation were used to investigate the anti-inflammatory activity of azadirachtin in mice. Zymosan-induced writhing and hot plate tests were employed to evaluate the antinociceptive activity. To explore putative mechanisms of action, the level of tumor necrosis factor-α in inflammatory tissue was measured and the effect induced by opioidergic and serotonergic antagonists was evaluated. Previous per os (p. o.) administration of azadirachtin (120 mg/kg) significantly reduced the acute paw edema induced by carrageenan. However, the concomitant increase of the paw concentration of tumor necrosis factor-α induced by this inflammatory stimulus was not reduced by azadirachtin. In addition to inhibiting the acute paw edema induced by carrageenan, azadirachtin (6, 60, and 120 mg/kg) inhibited the proliferative phase of the inflammatory response, as demonstrated by the reduced formation of fibrovascular tissue growth. Azadirachtin (120 mg/kg) also inhibited the nociceptive response in models of nociceptive (hot plate) and inflammatory (writhing induced by zymosan) pain. The activity of azadirachtin (120 mg/kg) in the model of nociceptive pain was attenuated by a nonselective opioid antagonist, naltrexone (10 mg/kg, i. p.), but not by a nonselective serotonergic antagonist, cyproheptadine. In conclusion, this study demonstrates the activity of azadirachtin in experimental models of nociceptive and inflammatory pain, and also in models of acute and chronic inflammation. Finally, multiple mechanisms, including the inhibition of the production of inflammatory mediators and activation of endogenous opioid pathways, may mediate azadirachtin activities in experimental models of inflammation and pain.


Asunto(s)
Analgésicos/farmacología , Antiinflamatorios/farmacología , Azadirachta/química , Edema/tratamiento farmacológico , Limoninas/farmacología , Extractos Vegetales/farmacología , Analgésicos/química , Analgésicos/aislamiento & purificación , Animales , Antiinflamatorios/química , Antiinflamatorios/aislamiento & purificación , Carragenina/efectos adversos , Modelos Animales de Enfermedad , Edema/inducido químicamente , Femenino , Inflamación/tratamiento farmacológico , Limoninas/química , Limoninas/aislamiento & purificación , Ratones , Nocicepción/efectos de los fármacos , Dolor/tratamiento farmacológico , Extractos Vegetales/química , Extractos Vegetales/aislamiento & purificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA