Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
J Neuroinflammation ; 17(1): 64, 2020 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-32070376

RESUMEN

BACKGROUND: Sepsis-associated acute brain inflammation, if unresolved, may cause chronic neuroinflammation and resultant neurodegenerative diseases. However, little is known how the transition from acute to chronic neuroinflammation, which is critical for the following progressive neurodegeneration, occurs in sepsis. The goal of this study was to investigate potential immune factors regulating the transition process using a widely used endotoxemia LPS mouse model. This model shows distinct acute and chronic phases of neuroinflammation and recapitulates many cardinal features of Parkinson's disease, thus, providing a unique opportunity for studying phase transition of neuroinflammation. METHODS: C57BL/6 J, NLRP3-/-, and IL-1R1-/- mice were employed. Mild and severe endotoxemia were produced by LPS ip injection at 1 or 5 mg/kg. Neuroinflammation in vitro and in vivo was assessed with proinflammatory cytokine expression by qPCR or ELISA and microglial activation by immunohistochemical analysis. Neurodegeneration was measured by manual and stereological counts of nigral dopaminergic neurons and immunohistochemical analysis of protein nitrosylation and α-synuclein phosphorylation. RESULTS: LPS-elicited initial increases in mouse brain mRNA levels of TNFα, IL-6, IL-1ß, and MCP-1, and nigral microglial activation were not dose-related. By contrast, the delayed increase in brain mature IL-1ß levels was dependent on LPS doses and protracted nigral microglial activation was only observed in high dose of LPS-treated mice. LPS-elicited increase in brain mature IL-1ß but not IL-1α level was NLRP3-dependent. After high dose LPS treatment, deficiency of NLRP3 or IL-1R1 did not prevent the initiation of acute neuroinflammation but abolished chronic neuroinflammation. Genetic or pharmacological inhibition of the NLRP3-IL-1ß axis repressed LPS-stimulated upregulation of chronic neuroinflammatory mediators including MHC-II, NOX2, and Mac1, and protected dopaminergic neurons. Ten months after LPS-elicited severe endotoxemia, nigral persisted microglial activation, elevated nitrosylated proteins and phosphorylated α-synuclein, and significant neuronal degeneration developed in wild-type mice but not in NLRP3-/- or IL-1R1-/- mice. CONCLUSIONS: This study uncovers a novel role of the NLRP3-IL-1ß signaling pathway in gauging the severity of sepsis-associated inflammation and determining whether acute neuroinflammation will resolve or transition to low grade chronic neuroinflammation. These findings also provide novel targets for developing therapy for severe systemic infection-related neurodegeneration.


Asunto(s)
Progresión de la Enfermedad , Mediadores de Inflamación/metabolismo , Interleucina-1beta/deficiencia , Proteína con Dominio Pirina 3 de la Familia NLR/deficiencia , Enfermedades Neurodegenerativas/metabolismo , Sepsis/metabolismo , Enfermedad Aguda , Animales , Células Cultivadas , Enfermedad Crónica , Lipopolisacáridos/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedades Neurodegenerativas/inducido químicamente , Sepsis/inducido químicamente
2.
Sci Rep ; 9(1): 10363, 2019 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-31316105

RESUMEN

Long-term peritoneal dialysis (PD) therapy leads to peritoneal inflammation and fibrosis. However, the mechanism underlying PD-related peritoneal inflammation and fibrosis remains unclear. NLRP3 inflammasome regulates the caspase-1-dependent release of interleukin-1ß and mediates inflammation in various diseases. Here, we investigated the role of NLRP3 inflammasome in a murine model of PD-related peritoneal fibrosis induced by methylglyoxal (MGO). Inflammasome-related proteins were upregulated in the peritoneum of MGO-treated mice. MGO induced parietal and visceral peritoneal fibrosis in wild-type mice, which was significantly reduced in mice deficient in NLRP3, ASC, and interleukin-1ß (IL-1ß). ASC deficiency reduced the expression of inflammatory cytokines and fibrotic factors, and the infiltration of macrophages. However, myeloid cell-specific ASC deficiency failed to inhibit MGO-induced peritoneal fibrosis. MGO caused hemorrhagic ascites, fibrin deposition, and plasminogen activator inhibitor-1 upregulation, but all of these manifestations were inhibited by ASC deficiency. Furthermore, in vitro experiments showed that MGO induced cell death via the generation of reactive oxygen species in vascular endothelial cells, which was inhibited by ASC deficiency. Our results showed that endothelial NLRP3 inflammasome contributes to PD-related peritoneal inflammation and fibrosis, and provide new insights into the mechanisms underlying the pathogenesis of this disorder.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/fisiología , Inflamasomas/fisiología , Interleucina-1beta/fisiología , Proteína con Dominio Pirina 3 de la Familia NLR/fisiología , Diálisis Peritoneal/efectos adversos , Fibrosis Peritoneal/etiología , Animales , Proteínas Adaptadoras de Señalización CARD/deficiencia , Proteínas Adaptadoras de Señalización CARD/genética , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Interleucina-1beta/deficiencia , Interleucina-1beta/genética , Leucocitos/patología , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/deficiencia , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Fibrosis Peritoneal/inducido químicamente , Fibrosis Peritoneal/patología , Piruvaldehído/toxicidad , Especies Reactivas de Oxígeno
3.
Biochem Pharmacol ; 155: 150-161, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29963998

RESUMEN

Interleukin (IL)-1 and IL-18 belong to the IL-1 family of ligands, and their receptors are members of the IL-1 receptor family. Both cytokines drive an extensive range of pro-inflammatory networks in many cell types using common signal transduction cascades. Anyway, differences in signaling pathways exist. With this aim in mind, we investigated by using transgenic mice the mechanisms through the simultaneous deficiency of both IL-1ß and IL-18 could be more protective compared to blocking the single cytokine IL-1ß or IL-18 during colitis. Colitis was provoked in mice by instillation of dinitrobenzene sulfonic acid (DNBS) in the colon. The results indicated that single knockout (KO) mice of IL-1ß or IL-18, and double KO mice of both IL-1ß and IL-18 were hyporesponsive to DNBS-induced colitis compared to wild type (WT) mice, in which double KO were less sensitive than single KO mice. Moreover, treatment with Anakinra (IL-1R antagonist) also ameliorated colitis, in views of macroscopic and histological alteration, infiltration of neutrophils or Th1 cells, oxidative and nitrosative stress. Anakinra more significantly reduced cyclooxygenase (COX-2) and nuclear factor (NF-κB) levels as well as IKB-α degradation compared to blocking IL-18. On the contrary, the absence of IL-18 reduced p-ERK and p-p38 mitogen-activated protein kinase (MAPKs) in a more significant way compared to blocking IL-1ß. Thus, the double KO increased the protective effects against colon inflammation maybe because different converging inflammatory pathways are being inhibited. In conclusion, the blocking of both IL-1ß and IL-18 function may be advantageous in the treatment of IBD or inflammatory diseases.


Asunto(s)
Colitis/inducido químicamente , Colitis/metabolismo , Dinitrofluorobenceno/análogos & derivados , Sistemas de Liberación de Medicamentos/métodos , Interleucina-18/deficiencia , Interleucina-1beta/deficiencia , Animales , Colitis/tratamiento farmacológico , Dinitrofluorobenceno/toxicidad , Proteína Antagonista del Receptor de Interleucina 1/administración & dosificación , Interleucina-18/antagonistas & inhibidores , Interleucina-1beta/antagonistas & inhibidores , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
4.
eNeuro ; 5(2)2018.
Artículo en Inglés | MEDLINE | ID: mdl-29662944

RESUMEN

Diffuse activation of interleukin-1 inflammatory cytokine signaling after traumatic brain injury (TBI) elicits progressive neurodegeneration and neuropsychiatric dysfunction, and thus represents a potential opportunity for therapeutic intervention. Although interleukin (IL)-1α and IL-1ß both activate the common type 1 IL-1 receptor (IL-1RI), they manifest distinct injury-specific roles in some models of neurodegeneration. Despite its potential relevance to treating patients with TBI, however, the individual contributions of IL-1α and IL-1ß to TBI-pathology have not been previously investigated. To address this need, we applied genetic and pharmacologic approaches in mice to dissect the individual contributions of IL-1α, IL-ß, and IL-1RI signaling to the pathophysiology of fluid percussion-mediated TBI, a model of mixed focal and diffuse TBI. IL-1RI ablation conferred a greater protective effect on brain cytokine expression and cognitive function after TBI than did individual IL-1α or IL-1ß ablation. This protective effect was recapitulated by treatment with the drug anakinra, a recombinant naturally occurring IL-1RI antagonist. Our data thus suggest that broad targeting of IL-1RI signaling is more likely to reduce neuroinflammation and preserve cognitive function after TBI than are approaches that individually target IL-1α or IL-1ß signaling.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Disfunción Cognitiva/prevención & control , Inflamación/prevención & control , Interleucina-1alfa/metabolismo , Interleucina-1beta/metabolismo , Transducción de Señal , Animales , Conducta Animal/efectos de los fármacos , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/inmunología , Lesiones Traumáticas del Encéfalo/metabolismo , Disfunción Cognitiva/etiología , Disfunción Cognitiva/inmunología , Disfunción Cognitiva/metabolismo , Modelos Animales de Enfermedad , Inflamación/etiología , Inflamación/inmunología , Inflamación/metabolismo , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Interleucina-1alfa/antagonistas & inhibidores , Interleucina-1alfa/deficiencia , Interleucina-1beta/antagonistas & inhibidores , Interleucina-1beta/deficiencia , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología
5.
Proc Natl Acad Sci U S A ; 115(16): E3673-E3681, 2018 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-29592953

RESUMEN

Metastases constitute the greatest causes of deaths from cancer. However, no effective therapeutic options currently exist for cancer patients with metastasis. Estrogen receptor ß (ERß), as a member of the nuclear receptor superfamily, shows potent tumor-suppressive activities in many cancers. To investigate whether modulation of ERß could serve as a therapeutic strategy for cancer metastasis, we examined whether the selective ERß agonist LY500307 could suppress lung metastasis of triple-negative breast cancer (TNBC) and melanoma. Mechanistically, while we observed that LY500307 potently induced cell death of cancer cells metastasized to lung in vivo, it does not mediate apoptosis of cancer cells in vitro, indicating that the cell death-inducing effects of LY500307 might be mediated by the tumor microenvironment. Pathological examination combined with flow cytometry assays indicated that LY500307 treatment induced significant infiltration of neutrophils in the metastatic niche. Functional experiments demonstrated that LY500307-treated cancer cells show chemotactic effects for neutrophils and that in vivo neutrophil depletion by Ly6G antibody administration could reverse the effects of LY500307-mediated metastasis suppression. RNA sequencing analysis showed that LY500307 could induce up-regulation of IL-1ß in TNBC and melanoma cells, which further triggered antitumor neutrophil chemotaxis. However, the therapeutic effects of LY500307 treatment for suppression of lung metastasis was attenuated in IL1B-/- murine models, due to failure to induce antitumor neutrophil infiltration in the metastatic niche. Collectively, our study demonstrated that pharmacological activation of ERß could augment innate immunity to suppress cancer metastatic colonization to lung, thus providing alternative therapeutic options for cancer patients with metastasis.


Asunto(s)
Antineoplásicos/farmacología , Benzopiranos/farmacología , Moduladores de los Receptores de Estrógeno/farmacología , Receptor beta de Estrógeno/agonistas , Inmunidad Innata/efectos de los fármacos , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/patología , Melanoma Experimental/secundario , Infiltración Neutrófila/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/terapia , Animales , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Benzopiranos/uso terapéutico , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Moduladores de los Receptores de Estrógeno/uso terapéutico , Estrógenos , Femenino , Interleucina-1beta/deficiencia , Interleucina-1beta/genética , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/terapia , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/terapia , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias Hormono-Dependientes/inmunología , Neoplasias Hormono-Dependientes/secundario , Neoplasias Hormono-Dependientes/terapia , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Organismos Libres de Patógenos Específicos , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
6.
Am J Physiol Regul Integr Comp Physiol ; 315(1): R90-R103, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29513560

RESUMEN

Skeletal muscle regeneration after injury is a complex process involving interactions between inflammatory microenvironments and satellite cells. Interleukin (IL)-1 is a key mediator of inflammatory responses and exerts pleiotropic impacts on various cell types. Thus, we aimed to investigate the role of IL-1 during skeletal muscle regeneration. We herein show that IL-1α/ß-double knockout (IL-1KO) mice exhibit delayed muscle regeneration after cardiotoxin (CTX) injection, characterized by delayed infiltrations of immune cells accompanied by suppressed local production of proinflammatory factors including IL-6 and delayed increase of paired box 7 (PAX7)-positive satellite cells postinjury compared with those of wild-type (WT) mice. A series of in vitro experiments using satellite cells obtained from the IL-1KO mice unexpectedly revealed that IL-1KO myoblasts have impairments in terms of both proliferation and differentiation, both of which were reversed by exogenous IL-1ß administration in culture. Intriguingly, the delay in myogenesis was not attributable to the myogenic transcriptional program since MyoD and myogenin were highly upregulated in IL-1KO cells, instead appearing, at least in part, to be due to dysregulation of cellular fusion events, possibly resulting from aberrant actin regulatory systems. We conclude that IL-1 plays a positive role in muscle regeneration by coordinating the initial interactions among inflammatory microenvironments and satellite cells. Our findings also provide compelling evidence that IL-1 is intimately engaged in regulating the fundamental function of myocytes.


Asunto(s)
Proliferación Celular , Interleucina-1alfa/metabolismo , Interleucina-1beta/metabolismo , Desarrollo de Músculos , Músculo Esquelético/metabolismo , Enfermedades Musculares/metabolismo , Regeneración , Células Satélite del Músculo Esquelético/metabolismo , Toxinas Biológicas , Animales , Modelos Animales de Enfermedad , Interleucina-1alfa/deficiencia , Interleucina-1alfa/genética , Interleucina-1beta/deficiencia , Interleucina-1beta/genética , Masculino , Ratones Endogámicos BALB C , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Enfermedades Musculares/inducido químicamente , Enfermedades Musculares/genética , Enfermedades Musculares/patología , Proteína MioD/genética , Proteína MioD/metabolismo , Miogenina/genética , Miogenina/metabolismo , Células Satélite del Músculo Esquelético/patología , Transducción de Señal , Nicho de Células Madre , Factores de Tiempo
7.
J Interferon Cytokine Res ; 38(3): 137-144, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29565745

RESUMEN

This study aimed to evaluate the role of eplerenone on the modulation of interleukin (IL)-1ß and IL-33/sST2 signaling pathway in an experimental model of left ventricular (LV) systolic dysfunction after acute myocardial infarction (MI). MI rats were randomly assigned to no treatment (MI group, n = 10), to receive eplerenone (Epl group, n = 10), or anakinra (Ana group, n = 10). LV function was assessed by echocardiography. IL-1ß, IL-33/sST2, and cardiac fibrosis biomarkers were analyzed by quantitative real-time reverse transcription polymerase chain reaction (PCR). Rats with MI showed significant reduction of LV systolic function, but treatment with eplerenone or anakinra improved left ventricular end-diastolic volume (LVEDV) and LVEDV/mass values. In the infarcted myocardium, compared with sham animals, the MI group had higher level of IL-33, sST2, and IL-1ß, as well as higher concentrations of markers of fibrosis and inflammation. Treatment with anakinra downregulated sST2 but with no effects on IL-33. Eplerenone reduced levels of sST2 and IL-1ß significantly. Both anakinra and eplerenone treatments were associated with lower levels of fibrosis and inflammatory markers. IL-1ß could induce expression of sST2, accelerating the progression of heart failure after acute MI. Eplerenone could improve LV function by reducing expression of IL-1ß and sST2.


Asunto(s)
Antihipertensivos/farmacología , Eplerenona/farmacología , Interleucina-1beta/metabolismo , Interleucina-33/metabolismo , Infarto del Miocardio/tratamiento farmacológico , Receptores de Interleucina-1/metabolismo , Transducción de Señal/efectos de los fármacos , Disfunción Ventricular Izquierda/tratamiento farmacológico , Enfermedad Aguda , Animales , Interleucina-1beta/deficiencia , Masculino , Infarto del Miocardio/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Interleucina-1/deficiencia , Disfunción Ventricular Izquierda/metabolismo
8.
Arterioscler Thromb Vasc Biol ; 38(4): 843-853, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29472233

RESUMEN

OBJECTIVE: Neutrophils promote experimental abdominal aortic aneurysm (AAA) formation via a mechanism that is independent from MMPs (matrix metalloproteinases). Recently, we reported a dominant role of IL (interleukin)-1ß in the formation of murine experimental AAAs. Here, the hypothesis that IL-1ß-induced neutrophil extracellular trap formation (NETosis) promotes AAA was tested. APPROACH AND RESULTS: NETs were identified through colocalized staining of neutrophil, Cit-H3 (citrullinated histone H3), and DNA, using immunohistochemistry. NETs were detected in human AAAs and were colocalized with IL-1ß. In vitro, IL-1RA attenuated IL-1ß-induced NETosis in human neutrophils. Mechanistically, IL-1ß treatment of isolated neutrophils induced nuclear localization of ceramide synthase 6 and synthesis of C16-ceramide, which was inhibited by IL-1RA or fumonisin B1, an inhibitor of ceramide synthesis. Furthermore, IL-1RA or fumonisin B1 attenuated IL1-ß-induced NETosis. In an experimental model of murine AAA, NETs were detected at a very early stage-day 3 of aneurysm induction. IL-1ß-knockout mice demonstrated significantly lower infiltration of neutrophils to aorta and were protected from AAA. Adoptive transfer of wild-type neutrophils promoted AAA formation in IL-1ß-knockout mice. Moreover, treatment of wild-type mice with Cl-amidine, an inhibitor NETosis, significantly attenuated AAA formation, whereas, treatment with deoxyribonuclease, a DNA digesting enzyme, had no effect on AAA formation. CONCLUSIONS: Altogether, the results suggest a dominant role of IL-1ß-induced NETosis in AAA formation.


Asunto(s)
Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/metabolismo , Trampas Extracelulares/metabolismo , Interleucina-1beta/metabolismo , Neutrófilos/metabolismo , Animales , Aorta Abdominal/efectos de los fármacos , Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/genética , Aneurisma de la Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/prevención & control , Ceramidas/metabolismo , Modelos Animales de Enfermedad , Trampas Extracelulares/efectos de los fármacos , Humanos , Procesamiento de Imagen Asistido por Computador/métodos , Interleucina-1beta/deficiencia , Interleucina-1beta/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Microscopía Fluorescente/métodos , Neutrófilos/efectos de los fármacos , Neutrófilos/patología , Neutrófilos/trasplante , Ornitina/análogos & derivados , Ornitina/farmacología , Receptores de Interleucina-1/metabolismo , Transducción de Señal , Esfingosina N-Aciltransferasa/metabolismo
9.
Radiat Res ; 189(3): 300-311, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29332538

RESUMEN

Lung exposure to radiation induces an injury response that includes the release of cytokines and chemotactic mediators; these signals recruit immune cells to execute inflammatory and wound-healing processes. However, radiation alters the pulmonary microenvironment, dysregulating the immune responses and preventing a return to homeostasis. Importantly, dysregulation is observed as a chronic inflammation, which can progress into pneumonitis and promote pulmonary fibrosis; inflammatory monocytes, which are bone marrow derived and express CCR2, have been shown to migrate into the lung after radiation exposure. Although the extent to which recruited inflammatory monocytes contribute to radiation-induced pulmonary fibrosis has not been fully investigated, we hypothesize that its pathogenesis is reliant on this population. The CC chemokine ligand, CCL2, is a chemotactic mediator responsible for trafficking of CCR2+ inflammatory cells into the lung. Therefore, the contribution of this mediator to fibrosis development was analyzed. Interleukin (IL)-1ß, a potent pro-inflammatory cytokine expressed during the radiation response, and its receptor, IL-1R1, were also evaluated. To this end, CCR2-/-, IL-1ß-/- and IL-1R1-/- chimeric mice were generated and exposed to 12.5 Gy thoracic radiation, and their response was compared to wild-type (C57BL/6) syngeneic controls. Fibrotic foci were observed in the periphery of the lungs of C57 syngeneic mice and CCR2-/- recipient mice that received C57 bone marrow (C57 > CCR2-/-) by 16 and 12 weeks after irradiation, respectively. In contrast, in the mice that had received bone marrow lacking CCR2 (CCR2-/- > C57 and CCR2-/- syngeneic mice), no pulmonary fibrosis was observed at 22 weeks postirradiation. This observation correlated with decreased numbers of infiltrating and interstitial macrophages compared to controls, as well as reduced proportions of pro-inflammatory Ly6C+ macrophages observed at 12-18 weeks postirradiation, suggesting that CCR2+ macrophages contribute to radiation-induced pulmonary fibrosis. Interestingly, reduced proportions of CD206+ lung macrophages were also present at these time points in CCR2-/- chimeric mice, regardless of donor bone marrow type, suggesting that the phenotype of resident subsets may be influenced by CCR2. Furthermore, chimeras, in which either IL-1ß was ablated from infiltrating cells or IL-1R1 from lung tissues, were also protected from fibrosis development, correlating with attenuated CCL2 production; these data suggest that IL-1ß may influence chemotactic signaling after irradiation. Overall, our data suggest that CCR2+ infiltrating monocyte-derived macrophages may play a critical role in the development of radiation-induced pulmonary fibrosis.


Asunto(s)
Monocitos/efectos de la radiación , Fibrosis Pulmonar/inmunología , Neumonitis por Radiación/inmunología , Animales , Relación Dosis-Respuesta en la Radiación , Femenino , Interleucina-1beta/deficiencia , Masculino , Ratones , Fenotipo , Fibrosis Pulmonar/metabolismo , Neumonitis por Radiación/metabolismo , Receptores CCR2/deficiencia , Receptores Tipo I de Interleucina-1/deficiencia
10.
Respir Res ; 19(1): 16, 2018 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-29361942

RESUMEN

BACKGROUND: Viral-induced asthma exacerbations, which exhibit both Th1-type neutrophilia and Th2-type inflammation, associate with secretion of Interleukin (IL)-1ß. IL-1ß induces neutrophilic inflammation. It may also increase Th2-type cytokine expression. We hypothesised that IL-1ß is causally involved in both Th1 and Th2 features of asthma exacerbations. This hypothesis is tested in our mouse model of viral stimulus-induced asthma exacerbation. METHOD: Wild-type (WT) and IL-1ß deficient (IL-1ß-/-) mice received house dust mite (HDM) or saline intranasally during three weeks followed by intranasal dsRNA (PolyI:C molecule known for its rhinovirus infection mimic) for three consecutive days to provoke exacerbation. Bronchoalveolar lavage fluid was analysed for inflammatory cells and total protein. Lung tissues were stained for neutrophilic inflammation and IL-33. Tissue homogenates were analysed for mRNA expression of Muc5ac, CXCL1/KC, TNF-α, CCL5, IL-25, TSLP, IL-33, IL-1ß, CCL11 and CCL2 using RT-qPCR. RESULTS: Expression of IL-1ß, neutrophil chemoattractants, CXCL1 and CCL5, the Th2-upstream cytokine IL-33, and Muc5ac were induced at exacerbation in WT mice and were significantly inhibited in IL-1ß-/- mice at exacerbation. Effects of HDM alone were not reduced in IL-1ß-deficient mice. CONCLUSION: Without being involved in the baseline HDM-induced allergic asthma, IL-1ß signalling was required to induce neutrophil chemotactic factors, IL-33, and Muc5ac expression at viral stimulus-induced exacerbation. We suggest that IL-1ß has a role both in neutrophilic and Th2 inflammation at viral-induced asthma exacerbations.


Asunto(s)
Asma/metabolismo , Modelos Animales de Enfermedad , Interleucina-1beta/deficiencia , Interleucina-33/biosíntesis , Neutrófilos/metabolismo , Pyroglyphidae , Animales , Asma/patología , Asma/virología , Expresión Génica , Interleucina-33/genética , Pulmón/metabolismo , Pulmón/patología , Pulmón/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/patología , Neutrófilos/virología , Rhinovirus
11.
Arterioscler Thromb Vasc Biol ; 38(2): 457-463, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29217508

RESUMEN

OBJECTIVE: Abdominal aortic aneurysms are inflammatory in nature and are associated with some risk factors that also lead to atherosclerotic occlusive disease, most notably smoking. The purpose of our study was to identify differential cytokine expression in patients with abdominal aortic aneurysm and those with atherosclerotic occlusive disease. Based on this analysis, we further explored and compared the mechanism of action of IL (interleukin)-1ß versus TNF-α (tumor necrosis factor-α) in abdominal aortic aneurysm formation. APPROACH AND RESULTS: IL-1ß was differentially expressed in human plasma with lower levels detected in patients with abdominal aortic aneurysm compared with matched atherosclerotic controls. We further explored its mechanism of action using a murine model and cell culture. Genetic deletion of IL-1ß and IL-1R did not inhibit aneurysm formation or decrease MMP (matrix metalloproteinase) expression. The effects of IL-1ß deletion on M1 macrophage polarization were compared with another proinflammatory cytokine, TNF-α. Bone marrow-derived macrophages from IL-1ß-/- and TNF-α-/- mice were polarized to an M1 phenotype. TNF-α deletion, but not IL-1ß deletion, inhibited M1 macrophage polarization. Infusion of M1 polarized TNF-α-/- macrophages inhibited aortic diameter growth; no inhibitory effect was seen in mice infused with M1 polarized IL-1ß-/- macrophages. CONCLUSIONS: Although IL-1ß is a proinflammatory cytokine, its effects on aneurysm formation and macrophage polarization differ from TNF-α. The differential effects of IL-1ß and TNF-α inhibition are related to M1/M2 macrophage polarization and this may account for the differences in clinical efficacy of IL-1ß and TNF-α antibody therapies in management of inflammatory diseases.


Asunto(s)
Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/metabolismo , Interleucina-1beta/metabolismo , Activación de Macrófagos , Macrófagos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Anciano , Animales , Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/patología , Estudios de Casos y Controles , Dilatación Patológica , Modelos Animales de Enfermedad , Femenino , Humanos , Interleucina-1beta/sangre , Interleucina-1beta/deficiencia , Interleucina-1beta/genética , Macrófagos/patología , Macrófagos/trasplante , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Fenotipo , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/deficiencia , Factor de Necrosis Tumoral alfa/genética
12.
PLoS One ; 12(5): e0176676, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28493895

RESUMEN

Cardiac glycosides such as digoxin are Na+/K+-ATPase inhibitors that are widely used for the treatment of chronic heart failure and cardiac arrhythmias; however, recent epidemiological studies have suggested a relationship between digoxin treatment and increased mortality. We previously showed that nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasomes, which regulate caspase-1-dependent interleukin (IL)-1ß release, mediate the sterile cardiovascular inflammation. Because the Na+/K+-ATPase is involved in inflammatory responses, we investigated the role of NLRP3 inflammasomes in the pathophysiology of cardiac glycoside-induced cardiac inflammation and dysfunction. The cardiac glycoside ouabain induced cardiac dysfunction and injury in wild-type mice primed with a low dose of lipopolysaccharide (LPS), although no cardiac dysfunction was observed in mice treated with either ouabain or LPS alone. Ouabain also induced cardiac inflammatory responses, such as macrophage infiltration and IL-1ß release, when mice were primed with LPS. These cardiac manifestations were all significantly attenuated in mice deficient in IL-1ß. Furthermore, deficiency of NLRP3 inflammasome components, NLRP3 and caspase-1, also attenuated ouabain-induced cardiac dysfunction and inflammation. In vitro experiments revealed that ouabain induced NLRP3 inflammasome activation as well as subsequent IL-1ß release from macrophages, and this activation was mediated by K+ efflux. Our findings demonstrate that cardiac glycosides promote cardiac inflammation and dysfunction through NLRP3 inflammasomes and provide new insights into the mechanisms underlying the adverse effects of cardiac glycosides.


Asunto(s)
Inflamasomas/metabolismo , Inflamación/patología , Inflamación/fisiopatología , Miocardio/metabolismo , Miocardio/patología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Ouabaína/efectos adversos , Animales , Transporte Biológico/efectos de los fármacos , Caspasa 1/metabolismo , Interleucina-1beta/deficiencia , Interleucina-1beta/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/patología , Ratones Endogámicos C57BL , Miocardio/enzimología , Potasio/metabolismo
13.
Sci Rep ; 6: 26489, 2016 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-27225830

RESUMEN

NLRP3 inflammasomes recognize non-microbial danger signals and induce release of proinflammatory cytokine interleukin (IL)-1ß, leading to sterile inflammation in cardiovascular disease. Because sterile inflammation is involved in doxorubicin (Dox)-induced cardiotoxicity, we investigated the role of NLRP3 inflammasomes in Dox-induced cardiotoxicity. Cardiac dysfunction and injury were induced by low-dose Dox (15 mg/kg) administration in NLRP3-deficient (NLRP3(-/-)) mice but not in wild-type (WT) and IL-1ß(-/-) mice, indicating that NLRP3 deficiency enhanced the susceptibility to Dox-induced cardiotoxicity independent of IL-1ß. Although the hearts of WT and NLRP3(-/-) mice showed no significant difference in inflammatory cell infiltration, macrophages were the predominant inflammatory cells in the hearts, and cardiac IL-10 production was decreased in Dox-treated NLRP3(-/-) mice. Bone marrow transplantation experiments showed that bone marrow-derived cells contributed to the exacerbation of Dox-induced cardiotoxicity in NLRP3(-/-) mice. In vitro experiments revealed that NLRP3 deficiency decreased IL-10 production in macrophages. Furthermore, adeno-associated virus-mediated IL-10 overexpression restored the exacerbation of cardiotoxicity in the NLRP3(-/-) mice. These results demonstrated that NLRP3 regulates macrophage IL-10 production and contributes to the pathophysiology of Dox-induced cardiotoxicity, which is independent of IL-1ß. Our findings identify a novel role of NLRP3 and provided new insights into the mechanisms underlying Dox-induced cardiotoxicity.


Asunto(s)
Doxorrubicina/toxicidad , Lesiones Cardíacas/inmunología , Interleucina-10/metabolismo , Interleucina-1beta/genética , Macrófagos/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Animales , Trasplante de Médula Ósea/efectos adversos , Cardiotoxicidad , Células Cultivadas , Modelos Animales de Enfermedad , Lesiones Cardíacas/inducido químicamente , Lesiones Cardíacas/genética , Humanos , Interleucina-1beta/deficiencia , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/deficiencia
14.
Arterioscler Thromb Vasc Biol ; 36(5): 886-97, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26941015

RESUMEN

OBJECTIVE: Kawasaki disease (KD) is the most common cause of acquired cardiac disease in US children. In addition to coronary artery abnormalities and aneurysms, it can be associated with systemic arterial aneurysms. We evaluated the development of systemic arterial dilatation and aneurysms, including abdominal aortic aneurysm (AAA) in the Lactobacillus casei cell-wall extract (LCWE)-induced KD vasculitis mouse model. METHODS AND RESULTS: We discovered that in addition to aortitis, coronary arteritis and myocarditis, the LCWE-induced KD mouse model is also associated with abdominal aorta dilatation and AAA, as well as renal and iliac artery aneurysms. AAA induced in KD mice was exclusively infrarenal, both fusiform and saccular, with intimal proliferation, myofibroblastic proliferation, break in the elastin layer, vascular smooth muscle cell loss, and inflammatory cell accumulation in the media and adventitia. Il1r(-/-), Il1a(-/-), and Il1b(-/-) mice were protected from KD associated AAA. Infiltrating CD11c(+) macrophages produced active caspase-1, and caspase-1 or NLRP3 deficiency inhibited AAA formation. Treatment with interleukin (IL)-1R antagonist (Anakinra), anti-IL-1α, or anti-IL-1ß mAb blocked LCWE-induced AAA formation. CONCLUSIONS: Similar to clinical KD, the LCWE-induced KD vasculitis mouse model can also be accompanied by AAA formation. Both IL-1α and IL-1ß play a key role, and use of an IL-1R blocking agent that inhibits both pathways may be a promising therapeutic target not only for KD coronary arteritis, but also for the other systemic arterial aneurysms including AAA that maybe seen in severe cases of KD. The LCWE-induced vasculitis model may also represent an alternative model for AAA disease.


Asunto(s)
Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/metabolismo , Interleucina-1alfa/metabolismo , Interleucina-1beta/metabolismo , Síndrome Mucocutáneo Linfonodular/complicaciones , Receptores Tipo I de Interleucina-1/metabolismo , Transducción de Señal , Animales , Aorta Abdominal/efectos de los fármacos , Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/genética , Aneurisma de la Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/prevención & control , Aortitis/genética , Aortitis/metabolismo , Aortitis/patología , Caspasa 1/deficiencia , Caspasa 1/genética , Proliferación Celular , Pared Celular , Dilatación Patológica , Modelos Animales de Enfermedad , Elastina/metabolismo , Femenino , Perfilación de la Expresión Génica , Genotipo , Humanos , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Interleucina-1alfa/deficiencia , Interleucina-1alfa/genética , Interleucina-1beta/deficiencia , Interleucina-1beta/genética , Lacticaseibacillus casei , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Síndrome Mucocutáneo Linfonodular/inducido químicamente , Síndrome Mucocutáneo Linfonodular/tratamiento farmacológico , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Proteína con Dominio Pirina 3 de la Familia NLR/deficiencia , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Fenotipo , Receptores Tipo I de Interleucina-1/deficiencia , Receptores Tipo I de Interleucina-1/genética , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
15.
Neurourol Urodyn ; 35(3): 377-81, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25557558

RESUMEN

AIMS: We investigated the relationship between IL-1ß and morphological and functional changes following partial bladder outlet obstruction (pBOO). METHODS: Female wild-type C57/BL6 mice (WT) and IL-1ß-/- mice (KO) were used. Animals were sacrificed either 1 or 3 weeks after pBOO or sham surgery, and their bladders were harvested to determine bladder weight, for RT-PCR to measure interleukin-1ß (IL-1ß), insulin growth factor-1 (IGF-1), and transforming growth factor-ß (TGF-ß) levels, and for histological analysis with Hematoxylin-Eosin (HE) staining. Cystometry was performed on conscious animals 3 weeks after surgery to evaluate urodynamic parameters. IGF-1 was also administered intraperitoneally to KO with pBOO, and bladder weight was then investigated. RESULTS: IL-1ß-mRNA levels were significantly higher in WT-pBOO than in WT-sham. IGF-1-mRNA and TGF-ß-mRNA levels were also significantly higher in WT-pBOO than in WT-sham; however, these increases were smaller in KO-pBOO than in WT-pBOO. Bladder weight was significantly higher in WT-pBOO than in WT-sham, while increases in bladder weight were significantly suppressed in KO-pBOO. HE staining revealed the thickened bladder wall in WT-pBOO, and this phenomenon was less in KO-pBOO than in WT-pBOO. Regarding the urodynamic parameters examined, micturition pressure and bladder capacity were significantly higher in WT-pBOO than in WT-sham, but remained unchanged in KO-pBOO. The administration of IGF-1 to KO-pBOO led to similar increases in bladder weight and the thickened bladder wall as those observed in WT-pBOO. CONCLUSION: IL-1ß has the potential to induce bladder remodeling and deteriorate urodynamic parameters in pBOO.


Asunto(s)
Proliferación Celular , Mediadores de Inflamación/metabolismo , Interleucina-1beta/metabolismo , Obstrucción del Cuello de la Vejiga Urinaria/metabolismo , Vejiga Urinaria/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Hipertrofia , Factor I del Crecimiento Similar a la Insulina/administración & dosificación , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Interleucina-1beta/deficiencia , Interleucina-1beta/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Presión , Transducción de Señal , Factores de Tiempo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Vejiga Urinaria/efectos de los fármacos , Vejiga Urinaria/patología , Vejiga Urinaria/fisiopatología , Obstrucción del Cuello de la Vejiga Urinaria/genética , Obstrucción del Cuello de la Vejiga Urinaria/patología , Obstrucción del Cuello de la Vejiga Urinaria/fisiopatología , Micción , Urodinámica
16.
J Gerontol A Biol Sci Med Sci ; 71(9): 1131-40, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26450946

RESUMEN

The infrapatellar fat pad (IFP) secretes inflammatory mediators in osteoarthritic knees, but the effect of aging on IFP inflammation is unknown. We tested the hypothesis that aging increases basal and interleukin-1ß (IL-1ß)-stimulated IFP inflammation in 10-, 20-, and 30-month-old male F344BN F1-hybrid rats. IFPs were cultured ex vivo for 24 hours and treated ±1ng/mL IL-1ß to simulate injury-induced inflammation. IFP inflammation was evaluated by measuring secreted cytokine concentrations and by quantitative expression of immunoregulatory and pro- and anti-adipogenic genes. With age, osteoarthritis pathology increased and IFP mass decreased. Although adipocyte size did not change with age, variation in adipocyte size was positively associated with synovial thickness independent of age whereas associations with cartilage damage were age dependent. In the absence of IL-1ß, aging was associated with a significant increase in IFP secretion of tumor necrosis factor α by 67% and IL-13 by 35% and a reduction in the expression of immunoregulatory M2 macrophage genes. However, following an IL-1ß challenge, adipogenesis markers decreased and pro- and anti-inflammatory cytokines increased independent of age. The lone exception was leptin, which decreased >70% with age. Thus, although aging promotes osteoarthritis risk by increasing basal inflammation, our findings also revealed a potentially protective effect of aging by decreasing IL-1ß-stimulated leptin production.


Asunto(s)
Tejido Adiposo/inmunología , Envejecimiento , Interleucina-1beta/inmunología , Articulación de la Rodilla/inmunología , Osteoartritis de la Rodilla/inmunología , Adipogénesis/inmunología , Tejido Adiposo/patología , Animales , Biomarcadores/sangre , Técnicas In Vitro , Inflamación/inmunología , Interleucina-13/inmunología , Interleucina-1beta/deficiencia , Articulación de la Rodilla/patología , Leptina/inmunología , Masculino , Osteoartritis de la Rodilla/diagnóstico , Valor Predictivo de las Pruebas , Ratas , Ratas Endogámicas F344 , Sensibilidad y Especificidad , Índice de Severidad de la Enfermedad , Factor de Necrosis Tumoral alfa/inmunología
17.
Cell Rep ; 13(9): 1922-36, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26655906

RESUMEN

Microbial pattern molecules in the intestine play immunoregulatory roles via diverse pattern recognition receptors. However, the role of the cytosolic DNA sensor AIM2 in the maintenance of intestinal homeostasis is unknown. Here, we show that Aim2(-/-) mice are highly susceptible to dextran sodium sulfate-induced colitis that is associated with microbial dysbiosis as represented by higher colonic burden of commensal Escherichia coli. Colonization of germ-free mice with Aim2(-/-) mouse microbiota leads to higher colitis susceptibility. In-depth investigation of AIM2-mediated host defense responses reveals that caspase-1 activation and IL-1ß and IL-18 production are compromised in Aim2(-/-) mouse colons, consistent with defective inflammasome function. Moreover, IL-18 infusion reduces E. coli burden as well as colitis susceptibility in Aim2(-/-) mice. Altered microbiota in inflammasome-defective mice correlate with reduced expression of several antimicrobial peptides in intestinal epithelial cells. Together, these findings implicate DNA sensing by AIM2 as a regulatory mechanism for maintaining intestinal homeostasis.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/metabolismo , Colon/metabolismo , Proteínas de Unión al ADN/genética , ADN/metabolismo , Animales , Caspasa 1/deficiencia , Caspasa 1/genética , Caspasa 1/metabolismo , Colitis/inducido químicamente , Colitis/metabolismo , Colitis/patología , Colon/microbiología , Citocinas/genética , Citocinas/metabolismo , ADN/química , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/metabolismo , Sulfato de Dextran/toxicidad , Susceptibilidad a Enfermedades , Disbiosis , Escherichia coli/patogenicidad , Heces/microbiología , Inflamasomas/metabolismo , Interleucina-18/deficiencia , Interleucina-18/genética , Interleucina-18/metabolismo , Interleucina-1beta/deficiencia , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microbiota , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo
18.
Eur J Immunol ; 45(2): 525-30, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25367678

RESUMEN

The immune system is implicated in a wide range of disorders affecting the brain and is, therefore, an attractive target for therapy. Interleukin-1 (IL-1) is a potent regulator of the innate immune system important for host defense but is also associated with injury and disease in the brain. Here, we show that IL-1 is a key mediator driving an innate immune response to inflammatory challenge in the mouse brain but is dispensable in extracerebral tissues including the lung and peritoneum. We also demonstrate that IL-1α is an important ligand contributing to the CNS dependence on IL-1 and that IL-1 derived from the CNS compartment (most likely microglia) is the major source driving this effect. These data reveal previously unknown tissue-specific requirements for IL-1 in driving innate immunity and suggest that IL-1-mediated inflammation in the brain could be selectively targeted without compromising systemic innate immune responses that are important for resistance to infection. This property could be exploited to mitigate injury- and disease-associated inflammation in the brain without increasing susceptibility to systemic infection, an important complication in several neurological disorders.


Asunto(s)
Encéfalo/inmunología , Encefalitis/inmunología , Interleucina-1alfa/genética , Interleucina-1beta/genética , Microglía/inmunología , Transducción de Señal/inmunología , Animales , Encéfalo/patología , Encefalitis/inducido químicamente , Encefalitis/genética , Encefalitis/patología , Regulación de la Expresión Génica , Inmunidad Innata , Inyecciones Intraventriculares , Interleucina-1alfa/deficiencia , Interleucina-1alfa/inmunología , Interleucina-1beta/deficiencia , Interleucina-1beta/inmunología , Lipopolisacáridos , Pulmón/inmunología , Ratones , Ratones Noqueados , Microglía/patología , Infiltración Neutrófila , Neutrófilos/inmunología , Neutrófilos/patología , Especificidad de Órganos , Peritoneo/inmunología
19.
Circulation ; 130(11 Suppl 1): S51-9, 2014 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-25200056

RESUMEN

BACKGROUND: Thoracic aortic aneurysms (TAAs) are common, but experimental TAA models are limited and the role of interleukin-1ß (IL-1ß) is undetermined. METHODS AND RESULTS: IL-1ß protein was measured in human TAAs and control aortas, and IL-1ß protein was increased ≈20-fold in human TAAs. To develop an experimental model of TAAs, 8- to 10-week-old male C57Bl/6 mice (wild type [WT]) underwent thoracotomy with application of periadventitial elastase (WT TAA) or saline (WT control; n=30 per group). Elastase treatment to thoracic aortas resulted in progressive dilation until day 14 with maximal dilation of 99.6±24.7% compared with 14.4±8.2% for WT saline control (P<0.0001). WT TAAs demonstrated elastin fragmentation, smooth muscle cell loss, macrophage infiltration, and increased IL-1ß expression. Next, TAAs were induced in mice deficient of IL-1ß (IL-1ß knockout) or IL-1 receptor (IL-1R knockout; n=10 each). Genetic deletion of IL-1ß and IL-1R significantly decreased thoracic aortic dilation (IL-1ß knockout=54.2±16.8% and IL-1R knockout=62.6±17.2% versus WT TAA=104.7±23.8%; P<0.001for both). IL-1ß knockout and IL-1R knockout aortas demonstrated preserved elastin and smooth muscle cells with fewer inflammatory cells. Correspondingly, IL-1ß and IL-1R knockout aortas had decreased inflammatory cytokine and matrix metalloproteinase 9 expression. Separately, WT mice pretreated with either IL-1R antagonist anakinra (100 mg/kg per day) or vehicle alone (control) underwent elastase treatment. Pretreatment of WT mice with anakinra attenuated TAA formation (control: 99.2±15.5% versus anakinra: 68.3±19.2%; P<0.005). Finally, to investigate treatment of small TAAs, WT mice were treated with anakinra 3 days after TAA induction. Anakinra treatment in WT mice with small TAAs reduced aortic dilation on day 14 (control treatment: 89.1±18.6% versus anakinra treatment: 59.7±25.7%; P=0.01). CONCLUSIONS: Periadventitial application of elastase to murine thoracic aortas reproducibly produced aneurysms with molecular and histological features consistent with TAA disease. Genetic and pharmacological inhibition of IL-1ß decreased TAA formation and progression, indicating that IL-1ß may be a potential target for TAA treatment.


Asunto(s)
Aneurisma de la Aorta Torácica/prevención & control , Proteína Antagonista del Receptor de Interleucina 1/uso terapéutico , Interleucina-1beta/antagonistas & inhibidores , Anciano , Animales , Aneurisma de la Aorta Torácica/inducido químicamente , Aneurisma de la Aorta Torácica/tratamiento farmacológico , Aneurisma de la Aorta Torácica/patología , Caspasa 1/fisiología , Comorbilidad , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Interleucina-1beta/deficiencia , Interleucina-1beta/genética , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Músculo Liso Vascular/patología , Elastasa Pancreática/toxicidad , Receptores de Interleucina-1/antagonistas & inhibidores , Receptores de Interleucina-1/deficiencia , Receptores de Interleucina-1/genética , Toracotomía
20.
Proc Natl Acad Sci U S A ; 111(34): 12492-7, 2014 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-25114230

RESUMEN

Oocyte endowment dwindles away during prepubertal and adult life until menopause occurs, and apoptosis has been identified as a central mechanism responsible for oocyte elimination. A few recent reports suggest that uncontrolled inflammation may adversely affect ovarian reserve. We tested the possible role of the proinflammatory cytokine IL-1 in the age-related exhaustion of ovarian reserve using IL-1α and IL-1ß-KO mice. IL-1α-KO mice showed a substantially higher pregnancy rate and litter size compared with WT mice at advanced age. The number of secondary and antral follicles was significantly higher in 2.5-mo-old IL-1α-KO ovaries compared with WT ovaries. Serum anti-Müllerian hormone, a putative marker of ovarian reserve, was markedly higher in IL-1α-KO mice from 2.5 mo onward, along with a greater ovarian response to gonadotropins. IL-1ß-KO mice displayed a comparable but more subtle prolongation of ovarian lifespan compared with IL-1α-KO mice. The protein and mRNA of both IL-1α and IL-1ß mice were localized within the developing follicles (oocytes and granulosa cells), and their ovarian mRNA levels increased with age. Molecular analysis revealed decreased apoptotic signaling [higher B-cell lymphoma 2 (BCL-2) and lower BCL-2-associated X protein levels], along with a marked attenuation in the expression of genes coding for the proinflammatory cytokines IL-1ß, IL-6, and TNF-α in ovaries of IL-1α-KO mice compared with WT mice. Taken together, IL-1 emerges as an important participant in the age-related exhaustion of ovarian reserve in mice, possibly by enhancing the expression of inflammatory genes and promoting apoptotic pathways.


Asunto(s)
Interleucina-1alfa/deficiencia , Interleucina-1beta/deficiencia , Ovario/fisiología , Envejecimiento , Animales , Hormona Antimülleriana/sangre , Apoptosis , Femenino , Expresión Génica , Mediadores de Inflamación/metabolismo , Interleucina-1alfa/genética , Interleucina-1alfa/fisiología , Interleucina-1beta/genética , Interleucina-1beta/fisiología , Tamaño de la Camada , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ovario/citología , Ovario/inmunología , Embarazo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de HFE/genética , Receptores de HFE/fisiología , Receptores Tipo I de Interleucina-1/deficiencia , Receptores Tipo I de Interleucina-1/genética , Receptores Tipo I de Interleucina-1/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...