Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
PLoS Pathog ; 16(10): e1008461, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33002089

RESUMEN

The induction of an interferon-mediated response is the first line of defense against pathogens such as viruses. Yet, the dynamics and extent of interferon alpha (IFNα)-induced antiviral genes vary remarkably and comprise three expression clusters: early, intermediate and late. By mathematical modeling based on time-resolved quantitative data, we identified mRNA stability as well as a negative regulatory loop as key mechanisms endogenously controlling the expression dynamics of IFNα-induced antiviral genes in hepatocytes. Guided by the mathematical model, we uncovered that this regulatory loop is mediated by the transcription factor IRF2 and showed that knock-down of IRF2 results in enhanced expression of early, intermediate and late IFNα-induced antiviral genes. Co-stimulation experiments with different pro-inflammatory cytokines revealed that this amplified expression dynamics of the early, intermediate and late IFNα-induced antiviral genes can also be achieved by co-application of IFNα and interleukin1 beta (IL1ß). Consistently, we found that IL1ß enhances IFNα-mediated repression of viral replication. Conversely, we observed that in IL1ß receptor knock-out mice replication of viruses sensitive to IFNα is increased. Thus, IL1ß is capable to potentiate IFNα-induced antiviral responses and could be exploited to improve antiviral therapies.


Asunto(s)
Regulación Viral de la Expresión Génica/efectos de los fármacos , Factor 2 Regulador del Interferón/metabolismo , Interferón-alfa/farmacología , Coriomeningitis Linfocítica/tratamiento farmacológico , Virus de la Coriomeningitis Linfocítica/efectos de los fármacos , Receptores Tipo I de Interleucina-1/fisiología , Replicación Viral/efectos de los fármacos , Animales , Antivirales/farmacología , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Hepatocitos/inmunología , Hepatocitos/virología , Humanos , Factor 2 Regulador del Interferón/genética , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/patología , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/aislamiento & purificación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estabilidad del ARN
2.
J Neurosci ; 40(47): 9103-9120, 2020 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-33051350

RESUMEN

Microglia are dynamic immunosurveillance cells in the CNS. Whether microglia are protective or pathologic is context dependent; the outcome varies as a function of time relative to the stimulus, activation state of neighboring cells in the microenvironment or within progression of a particular disease. Although brain microglia can be "primed" using bacterial lipopolysaccharide (LPS)/endotoxin, it is unknown whether LPS delivered systemically can also induce neuroprotective microglia in the spinal cord. Here, we show that serial systemic injections of LPS (1 mg/kg, i.p., daily) for 4 consecutive days (LPSx4) consistently elicit a reactive spinal cord microglia response marked by dramatic morphologic changes, increased production of IL-1, and enhanced proliferation without triggering leukocyte recruitment or overt neuropathology. Following LPSx4, reactive microglia frequently contact spinal cord endothelial cells. Targeted ablation or selective expression of IL-1 and IL-1 receptor (IL-1R) in either microglia or endothelia reveal that IL-1-dependent signaling between these cells mediates microglia activation. Using a mouse model of ischemic spinal cord injury in male and female mice, we show that preoperative LPSx4 provides complete protection from ischemia-induced neuron loss and hindlimb paralysis. Neuroprotection is partly reversed by either pharmacological elimination of microglia or selective removal of IL-1R in microglia or endothelia. These data indicate that spinal cord microglia are amenable to therapeutic reprogramming via systemic manipulation and that this potential can be harnessed to protect the spinal cord from injury.SIGNIFICANCE STATEMENT Data in this report indicate that a neuroprotective spinal cord microglia response can be triggered by daily systemic injections of LPS over a period of 4 d (LPSx4). The LPSx4 regimen induces morphologic transformation and enhances proliferation of spinal cord microglia without causing neuropathology. Using advanced transgenic mouse technology, we show that IL-1-dependent microglia-endothelia cross talk is necessary for eliciting this spinal cord microglia phenotype and also for conferring optimal protection to spinal motor neurons from ischemic spinal cord injury (ISCI). Collectively, these novel data show that it is possible to consistently elicit spinal cord microglia via systemic delivery of inflammogens to achieve a therapeutically effective neuroprotective response against ISCI.


Asunto(s)
Comunicación Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Interleucina-1/fisiología , Lipopolisacáridos/farmacología , Microglía/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Médula Espinal/efectos de los fármacos , Animales , Bromodesoxiuridina/farmacología , Células Endoteliales/metabolismo , Femenino , Interleucina-1/biosíntesis , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/patología , Parálisis/inducido químicamente , Receptores Tipo I de Interleucina-1/efectos de los fármacos , Receptores Tipo I de Interleucina-1/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Médula Espinal/metabolismo
3.
Am J Respir Cell Mol Biol ; 62(3): 300-309, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31499011

RESUMEN

Previous studies demonstrated spontaneous type 2 airway inflammation with eosinophilia in juvenile Scnn1b (sodium channel, non-voltage-gated 1, ß-subunit)-transgenic (Scnn1b-Tg) mice with muco-obstructive lung disease. IL-1 receptor (IL-1R) signaling has been implicated in allergen-driven airway disease; however, its role in eosinophilic inflammation in muco-obstructive lung disease remains unknown. In this study, we examined the role of IL-1R signaling in the development of airway eosinophilia and type 2 inflammation in juvenile Scnn1b-Tg mice. We determined effects of genetic deletion of Il1r1 (IL-1 receptor type I) on eosinophil counts, transcript levels of key type 2 cytokines, markers of eosinophil activation and apoptosis, and tissue morphology in lungs of Scnn1b-Tg mice at different time points during neonatal development. Furthermore, we measured endothelial surface expression of intercellular adhesion molecule 1 (ICAM-1), an integrin involved in eosinophil transendothelial migration, and determined effects of eosinophil depletion using an anti-IL-5 antibody on lung morphology. Lack of IL-1R reduced airway eosinophilia and structural lung damage, but it did not reduce concentrations of type 2 cytokines and associated eosinophil activation in Scnn1b-Tg mice. Structural lung damage in Scnn1b-Tg mice was also reduced by eosinophil depletion. Lack of IL-1R was associated with reduced expression of ICAM-1 on lung endothelial cells and reduced eosinophil counts in lungs from Scnn1b-Tg mice. We conclude that IL-1R signaling is implicated in airway eosinophilia independent of type 2 cytokines in juvenile Scnn1b-Tg mice. Our data suggest that IL-1R signaling may be relevant in the pathogenesis of eosinophilic airway inflammation in muco-obstructive lung diseases, which may be mediated in part by ICAM-1-dependent transmigration of eosinophils into the lungs.


Asunto(s)
Enfermedades Pulmonares Obstructivas/fisiopatología , Moco/metabolismo , Eosinofilia Pulmonar/fisiopatología , Receptores Tipo I de Interleucina-1/deficiencia , Envejecimiento/inmunología , Animales , Anticuerpos/farmacología , Anticuerpos/uso terapéutico , Apoptosis , Líquido del Lavado Bronquioalveolar/citología , Quimiotaxis de Leucocito , Citocinas/sangre , Citocinas/fisiología , Gránulos Citoplasmáticos/química , Gránulos Citoplasmáticos/ultraestructura , Células Endoteliales/metabolismo , Eosinófilos/efectos de los fármacos , Eosinófilos/inmunología , Eosinófilos/patología , Molécula 1 de Adhesión Intercelular/fisiología , Interleucina-5/inmunología , Enfermedades Pulmonares Obstructivas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Eosinofilia Pulmonar/tratamiento farmacológico , Eosinofilia Pulmonar/prevención & control , Receptores Tipo I de Interleucina-1/genética , Receptores Tipo I de Interleucina-1/fisiología , Transducción de Señal , Organismos Libres de Patógenos Específicos
4.
Mol Cancer Res ; 17(8): 1759-1773, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31164412

RESUMEN

Cancer development requires a favorable tissue microenvironment. By deleting Myd88 in keratinocytes or specific bone marrow subpopulations in oncogenic RAS-mediated skin carcinogenesis, we show that IL17 from infiltrating T cells and IκBζ signaling in keratinocytes are essential to produce a permissive microenvironment and tumor formation. Both normal and RAS-transformed keratinocytes respond to tumor promoters by activating canonical NF-κB and IκBζ signaling, releasing specific cytokines and chemokines that attract Th17 cells through MyD88-dependent signaling in T cells. The release of IL17 into the microenvironment elevates IκBζ in normal and RAS-transformed keratinocytes. Activation of IκBζ signaling is required for the expression of specific promoting factors induced by IL17 in normal keratinocytes and constitutively expressed in RAS-initiated keratinocytes. Deletion of Nfkbiz in keratinocytes impairs RAS-mediated benign tumor formation. Transcriptional profiling and gene set enrichment analysis of IκBζ-deficient RAS-initiated keratinocytes indicate that IκBζ signaling is common for RAS transformation of multiple epithelial cancers. Probing The Cancer Genome Atlas datasets using this transcriptional profile indicates that reduction of IκBζ signaling during cancer progression associates with poor prognosis in RAS-driven human cancers. IMPLICATIONS: The paradox that elevation of IκBζ and stimulation of IκBζ signaling through tumor extrinsic factors is required for RAS-mediated benign tumor formation while relative IκBζ expression is reduced in advanced cancers with poor prognosis implies that tumor cells switch from microenvironmental dependency early in carcinogenesis to cell-autonomous pathways during cancer progression.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Carcinogénesis/patología , Interleucina-17/metabolismo , Factor 88 de Diferenciación Mieloide/fisiología , Neoplasias Cutáneas/patología , Linfocitos T/metabolismo , Proteínas ras/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Carcinogénesis/genética , Carcinogénesis/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Interleucina-17/genética , Queratinocitos/metabolismo , Queratinocitos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/genética , FN-kappa B/metabolismo , Neoplasias Glandulares y Epiteliales/genética , Neoplasias Glandulares y Epiteliales/metabolismo , Neoplasias Glandulares y Epiteliales/patología , Receptores Tipo I de Interleucina-1/fisiología , Transducción de Señal , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Linfocitos T/patología , Microambiente Tumoral , Proteínas ras/genética
5.
PLoS Pathog ; 15(4): e1007744, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30978245

RESUMEN

Staphylococcus aureus is able to infect virtually all organ systems and is a frequently isolated etiologic agent of osteomyelitis, a common and debilitating invasive infection of bone. Treatment of osteomyelitis requires invasive surgical procedures and prolonged antibiotic therapy, yet is frequently unsuccessful due to extensive pathogen-induced bone damage that can limit antibiotic penetration and immune cell influx to the infectious focus. We previously established that S. aureus triggers profound alterations in bone remodeling in a murine model of osteomyelitis, in part through the production of osteolytic toxins. However, staphylococcal strains lacking osteolytic toxins still incite significant bone destruction, suggesting that host immune responses are also major drivers of pathologic bone remodeling during osteomyelitis. The objective of this study was to identify host immune pathways that contribute to antibacterial immunity during S. aureus osteomyelitis, and to define how these immune responses alter bone homeostasis and contribute to bone destruction. We specifically focused on the interleukin-1 receptor (IL-1R) and downstream adapter protein MyD88 given the prominent role of this signaling pathway in both antibacterial immunity and osteo-immunologic crosstalk. We discovered that while IL-1R signaling is necessary for local control of bacterial replication during osteomyelitis, it also contributes to bone loss during infection. Mechanistically, we demonstrate that S. aureus enhances osteoclastogenesis of myeloid precursors in vitro, and increases the abundance of osteoclasts residing on bone surfaces in vivo. This enhanced osteoclast abundance translates to trabecular bone loss, and is dependent on intact IL-1R signaling. Collectively, these data define IL-1R signaling as a critical component of the host response to S. aureus osteomyelitis, but also demonstrate that IL-1R-dependent immune responses trigger collateral bone damage through activation of osteoclast-mediated bone resorption.


Asunto(s)
Resorción Ósea/inmunología , Factor 88 de Diferenciación Mieloide/fisiología , Osteoclastos/inmunología , Osteomielitis/inmunología , Receptores Tipo I de Interleucina-1/fisiología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Animales , Resorción Ósea/metabolismo , Resorción Ósea/microbiología , Diferenciación Celular , Células Cultivadas , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoclastos/metabolismo , Osteoclastos/microbiología , Osteomielitis/metabolismo , Osteomielitis/microbiología , Transducción de Señal , Infecciones Estafilocócicas/metabolismo , Infecciones Estafilocócicas/microbiología
6.
J Hepatol ; 69(5): 1136-1144, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29886157

RESUMEN

BACKGROUND & AIMS: Biliary atresia (BA) results from a neonatal inflammatory and fibrosing obstruction of bile ducts of unknown etiology. Although the innate immune system has been linked to the virally induced mechanism of disease, the role of inflammasome-mediated epithelial injury remains largely undefined. Here, we hypothesized that disruption of the inflammasome suppresses the neonatal proinflammatory response and prevents experimental BA. METHODS: We determined the expression of key inflammasome-related genes in livers from infants at diagnosis of BA and in extrahepatic bile ducts (EHBDs) of neonatal mice after infection with rotavirus (RRV) immediately after birth. Then, we determined the impact of the wholesale inactivation of the genes encoding IL-1R1 (Il1r1-/-), NLRP3 (Nlrp3-/-) or caspase-1 (Casp1-/-) on epithelial injury and bile duct obstruction. RESULTS: IL1R1, NLRP3 and CASP1 mRNA increased significantly in human livers at the time of diagnosis, and in EHBDs of RRV-infected mice. In Il1r1-/- mice, the epithelial injury of EHBDs induced by RRV was suppressed, with dendritic cells unable to activate natural killer cells. A similar protection was observed in Nlrp3-/- mice, with decreased injury and inflammation of livers and EHBDs. Long-term survival was also improved. In contrast, the inactivation of the Casp1 gene had no impact on tissue injury, and all mice died. Tissue analyses in Il1r1-/- and Nlrp3-/- mice showed decreased populations of dendritic cells and natural killer cells and suppressed expression of type-1 cytokines and chemokines. CONCLUSIONS: Genes of the inflammasome are overexpressed at diagnosis of BA in humans and in the BA mouse model. In the experimental model, the targeted loss of IL-1R1 or NLRP3, but not of caspase-1, protected neonatal mice against RRV-induced bile duct obstruction. LAY SUMMARY: Biliary atresia is a severe inflammatory and obstructive disease of bile ducts occurring in infancy. Although the cause is unknown, activation of the innate and adaptive immune systems injures the bile duct epithelium. In this study we found that patients' livers had increased expression of inflammasome genes. Using mice engineered to inactivate individual inflammasome genes, the epithelial injury and bile duct obstruction were prevented by the loss of Il1r1 or Nlrp3, with a decreased activation of natural killer cells and expression of cytokines and chemokines. In contrast, the loss of Casp1 did not change the disease phenotype. Combined, the findings point to a differential role of inflammasome gene products in the pathogenic mechanisms of biliary atresia.


Asunto(s)
Atresia Biliar/etiología , Colestasis/etiología , Proteína con Dominio Pirina 3 de la Familia NLR/fisiología , Receptores Tipo I de Interleucina-1/fisiología , Animales , Animales Recién Nacidos , Atresia Biliar/patología , Caspasa 1/fisiología , Colestasis/patología , Células Dendríticas/inmunología , Epitelio/patología , Femenino , Humanos , Células Asesinas Naturales/inmunología , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos BALB C , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Receptores Tipo I de Interleucina-1/genética , Infecciones por Rotavirus/complicaciones
7.
Proc Natl Acad Sci U S A ; 115(21): E4843-E4852, 2018 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-29735654

RESUMEN

The nervous system maintains physiological homeostasis through reflex pathways that modulate organ function. This process begins when changes in the internal milieu (e.g., blood pressure, temperature, or pH) activate visceral sensory neurons that transmit action potentials along the vagus nerve to the brainstem. IL-1ß and TNF, inflammatory cytokines produced by immune cells during infection and injury, and other inflammatory mediators have been implicated in activating sensory action potentials in the vagus nerve. However, it remains unclear whether neural responses encode cytokine-specific information. Here we develop methods to isolate and decode specific neural signals to discriminate between two different cytokines. Nerve impulses recorded from the vagus nerve of mice exposed to IL-1ß and TNF were sorted into groups based on their shape and amplitude, and their respective firing rates were computed. This revealed sensory neural groups responding specifically to TNF and IL-1ß in a dose-dependent manner. These cytokine-mediated responses were subsequently decoded using a Naive Bayes algorithm that discriminated between no exposure and exposures to IL-1ß and TNF (mean successful identification rate 82.9 ± 17.8%, chance level 33%). Recordings obtained in IL-1 receptor-KO mice were devoid of IL-1ß-related signals but retained their responses to TNF. Genetic ablation of TRPV1 neurons attenuated the vagus neural signals mediated by IL-1ß, and distal lidocaine nerve block attenuated all vagus neural signals recorded. The results obtained in this study using the methodological framework suggest that cytokine-specific information is present in sensory neural signals within the vagus nerve.


Asunto(s)
Interleucina-1beta/farmacología , Receptores Tipo I de Interleucina-1/fisiología , Células Receptoras Sensoriales/fisiología , Canales Catiónicos TRPV/fisiología , Factor de Necrosis Tumoral alfa/farmacología , Nervio Vago/fisiología , Potenciales de Acción/efectos de los fármacos , Animales , Teorema de Bayes , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Células Receptoras Sensoriales/citología , Células Receptoras Sensoriales/efectos de los fármacos , Nervio Vago/citología , Nervio Vago/efectos de los fármacos
8.
Am J Physiol Lung Cell Mol Physiol ; 314(2): L225-L235, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28982735

RESUMEN

Acidic microenvironments commonly occur at sites of inflammation and bacterial infections. In the context of a Pseudomonas aeruginosa infection, we previously demonstrated that acidosis enhances the cellular proinflammatory interleukin (IL)-1ß response in vitro. However, how pH alterations affect in vivo IL-1ß responses and subsequent IL-1-driven inflammation during infection with P. aeruginosa is unclear. Here, we report that acidosis enhances in vivo IL-1ß production and downstream IL-1 receptor-dependent responses during infection with P. aeruginosa in models of acute pneumonia and peritonitis. Importantly, we demonstrate that infection with P. aeruginosa within an acidic environment leads to enhanced production of a subset of proinflammatory cytokines, including chemokine (C-X-C) motif ligand 1, IL-6, and chemokine (C-C motif) ligand 2, and increased neutrophil recruitment. Furthermore, with the use of IL-1 receptor type 1-deficient mice, we identify the contribution of the IL-1 signaling pathway to the acidosis-enhanced inflammatory response and pathology. These data provide insights into the potential benefit of pH regulation during bacterial infections to control disease progression and immunopathology.


Asunto(s)
Acidosis/fisiopatología , Inflamación/inmunología , Interleucina-1/farmacología , Neumonía Bacteriana/inmunología , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Receptores Tipo I de Interleucina-1/fisiología , Animales , Citocinas/metabolismo , Femenino , Inflamación/microbiología , Inflamación/patología , Mediadores de Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila , Neumonía Bacteriana/microbiología , Neumonía Bacteriana/patología , Infecciones por Pseudomonas/microbiología , Infecciones por Pseudomonas/patología
9.
Cancer Res ; 78(3): 695-705, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29217760

RESUMEN

Fatigue is the most common symptom of cancer at diagnosis, yet causes and effective treatments remain elusive. As tumors can be highly inflammatory, it is generally accepted that inflammation mediates cancer-related fatigue. However, evidence to support this assertion is mostly correlational. In this study, we directly tested the hypothesis that fatigue results from propagation of tumor-induced inflammation to the brain and activation of the central proinflammatory cytokine, IL1. The heterotopic syngeneic murine head and neck cancer model (mEER) caused systemic inflammation and increased expression of Il1b in the brain while inducing fatigue-like behaviors characterized by decreased voluntary wheel running and exploratory activity. Expression of Il1b in the brain was not associated with any alterations in motivation, measured by responding in a progressive ratio schedule of food reinforcement, depression-like behaviors, or energy balance. Decreased wheel running occurred prior to Il1b detection in the brain, when systemic inflammation was minimal. Furthermore, mice null for two components of IL1ß signaling, the type 1 IL1 receptor or the receptor adapter protein MyD88, were not protected from tumor-induced decreases in wheel running, despite attenuated cytokine action and expression. Behavioral and inflammatory analysis of four additional syngeneic tumor models revealed that tumors can induce fatigue regardless of their systemic or central nervous system inflammatory potential. Together, our results show that brain IL1 signaling is not necessary for tumor-related fatigue, dissociating this type of cancer sequela from systemic cytokine expression.Significance: These findings challenge the current understanding of fatigue in cancer patients, the most common and debilitating sequela associated with malignancy. Cancer Res; 78(3); 695-705. ©2017 AACR.


Asunto(s)
Encéfalo/patología , Fatiga/etiología , Neoplasias de Cabeza y Cuello/complicaciones , Inflamación/etiología , Interleucina-1beta/metabolismo , Factor 88 de Diferenciación Mieloide/fisiología , Receptores Tipo I de Interleucina-1/fisiología , Animales , Encéfalo/inmunología , Encéfalo/metabolismo , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Fatiga/metabolismo , Fatiga/patología , Inflamación/metabolismo , Inflamación/patología , Interleucina-1beta/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora , Transducción de Señal
10.
PLoS One ; 11(8): e0161539, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27536776

RESUMEN

In this study we sought to identify genetic factors associated with the presenting white blood cell (WBC) count in B-precursor acute lymphoblastic leukemia (BP-ALL). Using ETV6-RUNX1-positive BP-ALL patient samples, a homogeneous subtype, we identified 16 differentially expressed genes based on the presenting WBC count (< 50,000/cumm vs > 50,000). We further confirmed that IL1R1, BCAR3, KCNH2, PIR, and ZDHHC23 were differentially expressed in a larger cohort of ETV6-RUNX1-negative BP-ALL patient samples. Statistical analysis demonstrated that expression levels of these genes could accurately categorize high and low WBC count subjects using two independent patient sets, representing positive and negative ETV6-RUNX1 cases. Further studies in leukemia cell line models will better delineate the role of these genes in regulating the white blood cell count and potentially identify new therapeutic targets.


Asunto(s)
Recuento de Leucocitos , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/fisiología , Proteínas Portadoras/genética , Proteínas Portadoras/fisiología , Niño , Preescolar , Dioxigenasas , Canal de Potasio ERG1/genética , Canal de Potasio ERG1/fisiología , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Marcadores Genéticos/genética , Factores de Intercambio de Guanina Nucleótido , Humanos , Masculino , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Leucemia-Linfoma Linfoblástico de Células Precursoras B/inmunología , Mapas de Interacción de Proteínas/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Tipo I de Interleucina-1/genética , Receptores Tipo I de Interleucina-1/fisiología
11.
Diabetes Obes Metab ; 18(11): 1147-1151, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27350651

RESUMEN

Recently, glucagon-like peptide-1 (GLP-1) levels have been found to be increased in response to inflammatory stimuli, leading to insulin secretion and prevention of hyperglycaemia during endotoxemia in mice. In the present study, we assess the relevance of the other incretin hormone, glucose-dependent insulinotropic peptide (GIP), as a regulator of glucose metabolism under inflammatory conditions. We found that lipopolysaccharide (LPS) increased GIP secretion in a time- and dose-dependent manner in C57BL/6J mice. To elucidate the underlying mechanisms, mice were injected with inflammatory cytokines known to be released by LPS. Circulating GIP levels significantly increased in response to interleukin (IL)-1ß but not IL-6 or tumour necrosis factor (TNF)-α administration. Using respective knockout mice we found that LPS-mediated GIP secretion was selectively dependent on IL-1 signalling. To evaluate the functional relevance of inflammatory GIP secretion we pretreated mice with the GIP-receptor antagonist (Pro3)GIP. This blunted LPS-induced TNF-α and IL-6 secretion but did not affect LPS-induced insulin secretion or blood glucose-lowering. In conclusion, GIP provides a novel link between the immune system and the gut, with proinflammatory-immune modulatory function but minor glucose regulatory relevance in the context of acute endotoxemia.


Asunto(s)
Glucemia/metabolismo , Polipéptido Inhibidor Gástrico/metabolismo , Inflamación/inducido químicamente , Interleucina-1beta/fisiología , Lipopolisacáridos/farmacología , Receptores Tipo I de Interleucina-1/fisiología , Animales , Glucemia/efectos de los fármacos , Inflamación/metabolismo , Interleucina-6/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Tipo I de Interleucina-1/genética , Regulación hacia Arriba/efectos de los fármacos
12.
J Clin Invest ; 125(11): 4107-21, 2015 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-26436651

RESUMEN

Granulomatosis with polyangiitis (GPA) is a systemic necrotizing vasculitis that is associated with granulomatous inflammation and the presence of anti-neutrophil cytoplasmic antibodies (ANCAs) directed against proteinase 3 (PR3). We previously determined that PR3 on the surface of apoptotic neutrophils interferes with induction of antiinflammatory mechanisms following phagocytosis of these cells by macrophages. Here, we demonstrate that enzymatically active membrane-associated PR3 on apoptotic cells triggered secretion of inflammatory cytokines, including granulocyte CSF (G-CSF) and chemokines. This response required the IL-1R1/MyD88 signaling pathway and was dependent on the synthesis of NO, as macrophages from animals lacking these pathways did not exhibit a PR3-associated proinflammatory response. The PR3-induced microenvironment facilitated recruitment of inflammatory cells, such as macrophages, plasmacytoid DCs (pDCs), and neutrophils, which were observed in close proximity within granulomatous lesions in the lungs of GPA patients. In different murine models of apoptotic cell injection, the PR3-induced microenvironment instructed pDC-driven Th9/Th2 cell generation. Concomitant injection of anti-PR3 ANCAs with PR3-expressing apoptotic cells induced a Th17 response, revealing a GPA-specific mechanism of immune polarization. Accordingly, circulating CD4+ T cells from GPA patients had a skewed distribution of Th9/Th2/Th17. These results reveal that PR3 disrupts immune silencing associated with clearance of apoptotic neutrophils and provide insight into how PR3 and PR3-targeting ANCAs promote GPA pathophysiology.


Asunto(s)
Apoptosis/fisiología , Autoantígenos/fisiología , Granulomatosis con Poliangitis/inmunología , Mieloblastina/fisiología , Animales , Anticuerpos Anticitoplasma de Neutrófilos/inmunología , Autoantígenos/inmunología , Membrana Celular/enzimología , Microambiente Celular , Citocinas/metabolismo , Células Dendríticas/inmunología , Factor Estimulante de Colonias de Granulocitos/fisiología , Granulomatosis con Poliangitis/enzimología , Granulomatosis con Poliangitis/patología , Humanos , Pulmón/patología , Macrófagos/fisiología , Ratones , Ratones Endogámicos C57BL , Mieloblastina/biosíntesis , Mieloblastina/inmunología , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/fisiología , Neutrófilos/enzimología , Neutrófilos/inmunología , Neutrófilos/patología , Óxido Nítrico/fisiología , Peritonitis/inmunología , Peritonitis/patología , Fagocitosis , Receptores Tipo I de Interleucina-1/fisiología , Transducción de Señal , Subgrupos de Linfocitos T/inmunología
13.
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
14.
Brain Behav Immun ; 41: 218-31, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24907587

RESUMEN

Anxiety is one of the most commonly reported psychiatric conditions, but its pathogenesis is poorly understood. Ailments associated with activation of the innate immune system, however, are increasingly linked to anxiety disorders. In adult male mice, we found that adenosine doubled caspase-1 activity in brain by a pathway reliant on ATP-sensitive potassium (KATP) channels, protein kinase A (PKA) and the A2A adenosine receptor (AR). In addition, adenosine-dependent activation of caspase-1 increased interleukin (IL)-1ß in the brain by 2-fold. Peripheral administration of adenosine in wild-type (WT) mice led to a 2.3-fold increase in caspase-1 activity in the amygdala and to a 33% and 42% reduction in spontaneous locomotor activity and food intake, respectively, that were not observed in caspase-1 knockout (KO), IL-1 receptor type 1 (IL-1R1) KO and A2A AR KO mice or in mice administered a caspase-1 inhibitor centrally. Finally, adenosine administration increased anxiety-like behaviors in WT mice by 28% in the open field test and by 55% in the elevated zero-maze. Caspase-1 KO mice, IL-1R1 KO mice, A2A AR KO mice and WT mice treated with the KATP channel blocker, glyburide, were resistant to adenosine-induced anxiety-like behaviors. Thus, our results indicate that adenosine can act as an anxiogenic by activating caspase-1 and increasing IL-1ß in the brain.


Asunto(s)
Adenosina/toxicidad , Ansiedad/inducido químicamente , Encéfalo/metabolismo , Caspasa 1/fisiología , Interleucina-1beta/biosíntesis , Proteínas del Tejido Nervioso/fisiología , Receptor de Adenosina A2A/fisiología , Adenosina/farmacología , Amígdala del Cerebelo/metabolismo , Animales , Ansiedad/fisiopatología , Carbazoles/farmacología , Caspasa 1/deficiencia , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Activación Enzimática/efectos de los fármacos , Conducta Exploratoria/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Gliburida/farmacología , Interleucina-1beta/genética , Interleucina-1beta/fisiología , Transporte Iónico/efectos de los fármacos , Canales KATP/fisiología , Locomoción/efectos de los fármacos , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/deficiencia , Potasio/metabolismo , Pirroles/farmacología , Receptor de Adenosina A2A/deficiencia , Receptor de Adenosina A2A/efectos de los fármacos , Receptores Tipo I de Interleucina-1/deficiencia , Receptores Tipo I de Interleucina-1/fisiología
15.
Arterioscler Thromb Vasc Biol ; 34(3): 552-64, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24458711

RESUMEN

OBJECTIVE: Interleukin 1 Receptor 1 (IL1R1) and its ligand, IL1ß, are upregulated in cardiovascular disease, obesity, and infection. Previously, we reported a higher level of IL1R1 transcripts in platelets from obese individuals of the Framingham Heart Study (FHS), but its functional effect in platelets has never been described. Additionally, IL1ß levels are increased in atherosclerotic plaques and in bacterial infections. The aim of this work is to determine whether IL1ß, through IL1R1, can activate platelets and megakaryocytes to promote atherothrombosis. APPROACH AND RESULTS: We found that IL1ß-related genes from platelets, as measured in 1819 FHS participants, were associated with increased body mass index, and a direct relationship was shown in wild-type mice fed a high-fat diet. Mechanistically, IL1ß activated nuclear factor-κB and mitogen-activated protein kinase signaling pathways in megakaryocytes. IL1ß, through IL1R1, increased ploidy of megakaryocytes to 64+ N by 2-fold over control. IL1ß increased agonist-induced platelet aggregation by 1.2-fold with thrombin and 4.2-fold with collagen. IL1ß increased adhesion to both collagen and fibrinogen, and heterotypic aggregation by 1.9-fold over resting. High fat diet-enhanced platelet adhesion was absent in IL1R1(-/-) mice. Wild-type mice infected with Porphyromonas gingivalis had circulating heterotypic aggregates (1.5-fold more than control at 24 hours and 6.2-fold more at 6 weeks) that were absent in infected IL1R1(-/-) and IL1ß(-/-) mice. CONCLUSIONS: In summary, IL1R1- and IL1ß-related transcripts are elevated in the setting of obesity. IL1R1/IL1ß augment both megakaryocyte and platelet functions, thereby promoting a prothrombotic environment during infection and obesity; potentially contributing to the development of atherothrombotic disease.


Asunto(s)
Inflamación/patología , Interleucina-1beta/fisiología , Megacariocitos/citología , Obesidad/sangre , Activación Plaquetaria/fisiología , Receptores Tipo I de Interleucina-1/fisiología , Transcripción Genética/fisiología , Animales , Aterosclerosis/etiología , Infecciones por Bacteroidaceae/sangre , Infecciones por Bacteroidaceae/patología , Línea Celular , Colágeno/farmacología , Grasas de la Dieta/toxicidad , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Humanos , Imidazoles/farmacología , Inflamación/etiología , Inflamación/genética , Interleucina-1beta/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Ratones Noqueados , FN-kappa B/metabolismo , Obesidad/complicaciones , Obesidad/genética , Fosforilación/efectos de los fármacos , Activación Plaquetaria/efectos de los fármacos , Adhesividad Plaquetaria/efectos de los fármacos , Adhesividad Plaquetaria/fisiología , Porphyromonas gingivalis , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piridinas/farmacología , Receptores Tipo I de Interleucina-1/deficiencia , Receptores Tipo I de Interleucina-1/genética , Trombina/farmacología , Transcripción Genética/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
16.
Vascul Pharmacol ; 60(1): 1-7, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23806284

RESUMEN

The cardiac fibroblast (CF) is a multifunctional and heterogeneous cell type that plays an essential role in regulating cardiac development, structure and function. Following myocardial infarction (MI), the myocardium undergoes complex structural remodelling in an attempt to repair the damaged tissue and overcome the loss of function induced by ischemia/reperfusion injury. Evidence is emerging that CF play critical roles in all stages of post-MI remodelling, including the initial inflammatory phase that is triggered in response to myocardial damage. CF are particularly responsive to the proinflammatory cytokine interleukin-1 (IL-1) whose levels are rapidly induced in the myocardium after MI. Studies from our laboratory in recent years have sought to evaluate the functional effects of IL-1 on human CF function and to determine the underlying molecular mechanisms. This review summarises these data and sets it in the context of post-MI cardiac remodelling, identifying the fibroblast as a potential therapeutic target for reducing adverse cardiac remodelling and its devastating consequences.


Asunto(s)
Fibroblastos/efectos de los fármacos , Interleucina-1/farmacología , Infarto del Miocardio/patología , Remodelación Ventricular , Animales , Proliferación Celular , Matriz Extracelular/metabolismo , Fibroblastos/fisiología , Humanos , Receptores Tipo I de Interleucina-1/fisiología , Transducción de Señal
17.
Brain Behav Immun ; 35: 135-43, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24060584

RESUMEN

Challenges experienced in early life cause an enduring phenotypical shift of immune cells towards a sensitised state that may lead to an exacerbated reaction later in life and contribute to increased vulnerability to neurological diseases. Peripheral and central inflammation may affect neuronal function through cytokines such as IL-1. The extent to which an early life challenge induces long-term alteration of immune receptors organization in neurons has not been shown. We investigated whether a single episode of maternal deprivation (MD) on post-natal day (PND) 9 affects: (i) the synapse distribution of IL-1RI together with subunits of NMDA and AMPA receptors; and (ii) the interactions between IL-1RI and the GluN2B subunit of the NMDAR in the long-term, at PND 45. MD increased IL-1RI levels and IL-1RI interactions with GluN2B at the synapse of male hippocampal neurons, without affecting the total number of IL-1RI or NMDAR subunits. Although GluN2B and GluN2A were slightly but not significantly changed at the synapse, their ratio was significantly decreased in the hippocampus of the male rats who had experienced MD; the levels of the GluA1 and GluA2 subunits of the AMPAR were also decreased. These changes were not observed immediately after the MD episode. None of the observed alterations occurred in the hippocampus of the females or in the prefrontal cortex of either sex. These data reveal a long-term, sex-dependent modification in receptor organisation at the hippocampal post-synapses following MD. We suggest that this effect might contribute to priming hippocampal synapses to the action of IL-1ß.


Asunto(s)
Hipocampo/inmunología , Privación Materna , Receptores Tipo I de Interleucina-1/fisiología , Sinapsis/inmunología , Animales , Western Blotting , Femenino , Hipocampo/química , Hipocampo/fisiología , Inmunoprecipitación , Interleucina-1beta/análisis , Masculino , Ratas , Ratas Wistar , Factores Sexuales , Fracciones Subcelulares/metabolismo , Sinapsis/fisiología
18.
Am J Physiol Endocrinol Metab ; 305(1): E15-21, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23592480

RESUMEN

The proinflammatory cytokine interleukin-1 (IL-1) signals through IL-1 receptor type I (IL-1RI) and induces osteoclastogenesis and bone resorption mainly during pathological conditions. Little is known about the effect of excess or absence of IL-1 signaling on the physiological development of the growth plate and bone. In this study, we examine growth plate morphology, bone structure, and mechanical properties as well as osteoclast number in IL-1RI knockout mice to evaluate the role of IL-1RI in the normal development of the growth plate and bone. We show for the first time that IL-1RI knockout mice have narrower growth plates due to a smaller hypertrophic zone, suggesting a role for this cytokine in hypertrophic differentiation, together with higher proteoglycan content. The bones of theses mice exhibit higher trabecular and cortical mass, increased mineral density, and superior mechanical properties. In addition, IL-1RI knockout mice have significantly reduced osteoclast numbers in the chondro-osseous junction, trabecular bone, and cortical bone. These results suggest that IL-1RI is involved in normal growth plate development and ECM homeostasis and that it is significant in the physiological process of bone modeling.


Asunto(s)
Remodelación Ósea/fisiología , Placa de Crecimiento/crecimiento & desarrollo , Placa de Crecimiento/fisiología , Receptores Tipo I de Interleucina-1/fisiología , Transducción de Señal/fisiología , Animales , Placa de Crecimiento/diagnóstico por imagen , Homeostasis/fisiología , Interleucina-1beta/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoclastos/citología , Osteoclastos/fisiología , Proteoglicanos/metabolismo , Radiografía , Receptores Tipo I de Interleucina-1/genética , Receptores Tipo I de Interleucina-1/metabolismo , Tibia/diagnóstico por imagen , Tibia/crecimiento & desarrollo , Tibia/fisiología
19.
J Exp Med ; 210(3): 503-16, 2013 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-23460727

RESUMEN

Infections of the central nervous system (CNS) with cytopathic viruses require efficient T cell responses to promote viral clearance, limit immunopathology, and enhance survival. We found that IL-1R1 is critical for effector T cell reactivation and limits inflammation within the CNS during murine West Nile virus (WNV) encephalitis. WNV-infected IL-1R1(-/-) mice display intact adaptive immunity in the periphery but succumb to WNV infection caused by loss of virologic control in the CNS with depressed local Th1 cytokine responses, despite parenchymal entry of virus-specific CD8(+) T cells. Ex vivo analysis of CD4(+) T cells from WNV-infected CNS of IL-1R1(-/-) mice revealed impaired effector responses, whereas CD8(+) T cells revealed no cell intrinsic defects in response to WNV antigen. WNV-infected, IL-1R1(-/-) mice also exhibited decreased activation of CNS CD11c(+)CD11b(-)CD103(+) and CD11c(+)CD11b(-)CD8α(+)Dec-205(+) cells with reduced up-regulation of the co-stimulatory molecules CD80, CD86, and CD68. Adoptive transfer of wild-type CD11c-EYFP(+) cells from WNV-infected CNS into WNV-infected IL-1R1(-/-) mice trafficked into the CNS restored T cell functions and improved survival from otherwise lethal infection. These data indicate that IL-1R1 signaling promotes virologic control during WNV infection specifically within the CNS via modulation of CD11c(+) cell-mediated T cell reactivation at this site.


Asunto(s)
Encéfalo/inmunología , Células Dendríticas/inmunología , Encefalitis Viral/inmunología , Activación de Linfocitos , Receptores Tipo I de Interleucina-1/fisiología , Linfocitos T/inmunología , Fiebre del Nilo Occidental/inmunología , Traslado Adoptivo , Animales , Antígeno CD11c/análisis , Humanos , Interleucina-1/fisiología , Ratones , Ratones Endogámicos C57BL , Transducción de Señal
20.
J Clin Invest ; 122(10): 3476-89, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22945633

RESUMEN

Alcoholic liver disease (ALD) is characterized by steatosis and upregulation of proinflammatory cytokines, including IL-1ß. IL-1ß, type I IL-1 receptor (IL-1R1), and IL-1 receptor antagonist (IL-1Ra) are all important regulators of the IL-1 signaling complex, which plays a role in inflammation. Furthermore, IL-1ß maturation is dependent on caspase-1 (Casp-1). Using IL-1Ra-treated mice as well as 3 mouse models deficient in regulators of IL-1ß activation (Casp-1 and ASC) or signaling (IL-1R1), we found that IL-1ß signaling is required for the development of alcohol-induced liver steatosis, inflammation, and injury. Increased IL-1ß was due to upregulation of Casp-1 activity and inflammasome activation. The pathogenic role of IL-1 signaling in ALD was attributable to the activation of the inflammasome in BM-derived Kupffer cells. Importantly, in vivo intervention with a recombinant IL-1Ra blocked IL-1 signaling and markedly attenuated alcohol-induced liver inflammation, steatosis, and damage. Furthermore, physiological doses of IL-1ß induced steatosis, increased the inflammatory and prosteatotic chemokine MCP-1 in hepatocytes, and augmented TLR4-dependent upregulation of inflammatory signaling in macrophages. In conclusion, we demonstrated that Casp-1-dependent upregulation of IL-1ß and signaling mediated by IL-1R1 are crucial in ALD pathogenesis. Our findings suggest a potential role of IL-1R1 inhibition in the treatment of ALD.


Asunto(s)
Hígado Graso Alcohólico/tratamiento farmacológico , Inflamasomas/fisiología , Proteína Antagonista del Receptor de Interleucina 1/uso terapéutico , Animales , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/genética , Caspasa 1/fisiología , Quimiocina CCL2/biosíntesis , Quimiocina CCL2/genética , Evaluación Preclínica de Medicamentos , Etanol/toxicidad , Hígado Graso Alcohólico/etiología , Femenino , Hepatocitos/metabolismo , Interleucina-1alfa/biosíntesis , Interleucina-1alfa/sangre , Interleucina-1beta/biosíntesis , Interleucina-1beta/sangre , Interleucina-1beta/fisiología , Interleucina-1beta/toxicidad , Macrófagos del Hígado/patología , Cirrosis Hepática/etiología , Cirrosis Hepática/prevención & control , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Receptores Tipo I de Interleucina-1/deficiencia , Receptores Tipo I de Interleucina-1/fisiología , Proteínas Recombinantes/uso terapéutico , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/fisiología , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...