Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 19(6): e1011455, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37347786

RESUMEN

XIAP is an endogenous inhibitor of cell death and inactivating mutations of XIAP are responsible for X-linked lymphoproliferative disease (XLP-2) and primary immunodeficiency, but the mechanism(s) behind these contradictory outcomes have been unclear. We report that during infection of macrophages and dendritic cells with various intracellular bacteria, XIAP restricts cell death and secretion of IL-1ß but promotes increased activation of NFκB and JNK which results in elevated secretion of IL-6 and IL-10. Poor secretion of IL-6 by Xiap-deficient antigen presenting cells leads to poor expansion of recently activated CD8 T cells during the priming phase of the response. On the other hand, Xiap-deficient CD8 T cells displayed increased proliferation and effector function during the priming phase but underwent enhanced contraction subsequently. Xiap-deficient CD8 T cells underwent skewed differentiation towards short lived effectors which resulted in poor generation of memory. Consequently Xiap-deficient CD8 T cells failed to provide effective control of bacterial infection during re-challenge. These results reveal the temporal impact of XIAP in promoting the fitness of activated CD8 T cells through cell extrinsic and intrinsic mechanisms and provide a mechanistic explanation of the phenotype observed in XLP-2 patients.


Asunto(s)
Interleucina-6 , Trastornos Linfoproliferativos , Humanos , Muerte Celular , Trastornos Linfoproliferativos/genética , Macrófagos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Memoria Inmunológica , Proteína Inhibidora de la Apoptosis Ligada a X/genética , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo
2.
Nat Immunol ; 13(10): 954-62, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22922364

RESUMEN

Salmonella enterica serovar Typhimurium (S. Typhimurium) is a virulent pathogen that induces rapid host death. Here we observed that host survival after infection with S. Typhimurium was enhanced in the absence of type I interferon signaling, with improved survival of mice deficient in the receptor for type I interferons (Ifnar1(-/-) mice) that was attributed to macrophages. Although there was no impairment in cytokine expression or inflammasome activation in Ifnar1(-/-) macrophages, they were highly resistant to S. Typhimurium-induced cell death. Specific inhibition of the kinase RIP1 or knockdown of the gene encoding the kinase RIP3 prevented the death of wild-type macrophages, which indicated that necroptosis was a mechanism of cell death. Finally, RIP3-deficient macrophages, which cannot undergo necroptosis, had similarly less death and enhanced control of S. Typhimurium in vivo. Thus, we propose that S. Typhimurium induces the production of type I interferon, which drives necroptosis of macrophages and allows them to evade the immune response.


Asunto(s)
Proteínas Activadoras de GTPasa/metabolismo , Interferón Tipo I/metabolismo , Macrófagos/fisiología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Salmonelosis Animal/inmunología , Salmonella typhimurium/patogenicidad , Animales , Apoptosis , Proteínas Activadoras de GTPasa/antagonistas & inhibidores , Evasión Inmune , Inflamasomas , Interferón Tipo I/inmunología , Activación de Macrófagos , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/antagonistas & inhibidores , Proteína Serina-Treonina Quinasas de Interacción con Receptores/deficiencia , Salmonelosis Animal/metabolismo , Salmonelosis Animal/microbiología , Salmonella typhimurium/inmunología , Transducción de Señal
3.
J Biol Chem ; 298(1): 101461, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34864057

RESUMEN

Inflammasome signaling results in cell death and release of cytokines from the IL-1 family, which facilitates control over an infection. However, some pathogens such as Salmonella typhimurium (ST) activate various innate immune signaling pathways, including inflammasomes, yet evade these cell death mechanisms, resulting in a chronic infection. Here we investigated inflammasome signaling induced by acute and chronic isolates of ST obtained from different organs. We show that ST isolated from infected mice during the acute phase displays an increased potential to activate inflammasome signaling, which then undergoes a protracted decline during the chronic phase of infection. This decline in inflammasome signaling was associated with reduced expression of virulence factors, including flagella and the Salmonella pathogenicity island I genes. This reduction in cell death of macrophages induced by chronic isolates had the greatest impact on the NLRP3 inflammasome, which correlated with a reduction in caspase-1 activation. Furthermore, rapid cell death induced by Casp-1/11 by ST in macrophages limited the subsequent activation of cell death cascade proteins Casp-8, RipK1, RipK3, and MLKL to prevent the activation of alternative forms of cell death. We observed that the lack of the ability to induce cell death conferred a competitive fitness advantage to ST only during the acute phase of infection. Finally, we show that the chronic isolates displayed a significant attenuation in their ability to infect mice through the oral route. These results reveal that ST adapts during chronic infection by circumventing inflammasome recognition to promote the survival of both the host and the pathogen.


Asunto(s)
Inflamasomas , Macrófagos , Proteína con Dominio Pirina 3 de la Familia NLR , Infecciones por Salmonella , Salmonella typhimurium , Animales , Caspasa 1/genética , Caspasa 1/metabolismo , Interacciones Huésped-Patógeno/inmunología , Inflamasomas/inmunología , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Infecciones por Salmonella/inmunología , Infecciones por Salmonella/microbiología , Salmonella typhimurium/inmunología , Salmonella typhimurium/aislamiento & purificación
4.
J Biol Chem ; 298(10): 102386, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35985421

RESUMEN

The intestine-specific transcription factor Cdx2 is essential for intestinal homeostasis and has been implicated in the pathogenesis of disorders including inflammatory bowel disease. However, the mechanism by which Cdx2 influences intestinal disease is not clear. Here, we present evidence supporting a novel Cdx2-TRIM31-NLRP3 (NLR family, pyrin domain containing 3) signaling pathway, which may represent a mechanistic means by which Cdx2 impacts intestinal inflammation. We found that conditional loss of Cdx function resulted in an increase in proinflammatory cytokines, including tumor necrosis factor alpha, interleukin (IL)-1ß, and IL-6, in the mouse colon. We further show that TRIM31, which encodes a suppressor of NLRP3 (a central component of the NLRP3 inflammasome complex) is a novel Cdx2 target gene and is attenuated in the colon of Cdx conditional mutants. Consistent with this, we found that attenuation of TRIM31 in Cdx mutant intestine occurs concomitant with elevated levels of NLRP3 and an increase in inflammasome products. We demonstrate that specific inhibition of NLRP3 activity significantly reduced IL-1ß and IL-6 levels and extended the life span of Cdx conditional mutants, reflecting the therapeutic potential of targeting NLRP3. Tumor necrosis factor-alpha levels were also induced independent of NLRP3, potentially via elevated activity of the proinflammatory NF-κB signaling pathway in Cdx mutants. Finally, in silico analysis of ulcerative colitis patients revealed attenuation of CDX2 and TRIM31 expression coincident with enhanced expression of proinflammatory cytokines. We conclude that the novel Cdx2-TRIM31-NLRP3 signaling pathway promotes proinflammatory cytokine expression, and its inhibition may have therapeutic potential in human intestinal diseases.


Asunto(s)
Factor de Transcripción CDX2 , Inflamasomas , Enfermedades Inflamatorias del Intestino , Proteína con Dominio Pirina 3 de la Familia NLR , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas , Animales , Humanos , Ratones , Factor de Transcripción CDX2/genética , Factor de Transcripción CDX2/metabolismo , Citocinas/metabolismo , Homeostasis , Inflamasomas/genética , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/genética , Intestinos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas de Motivos Tripartitos/genética , Proteínas de Motivos Tripartitos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo
5.
J Biol Chem ; 295(14): 4661-4672, 2020 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-32094226

RESUMEN

The necrosome is a protein complex required for signaling in cells that results in necroptosis, which is also dependent on tumor necrosis factor receptor (TNF-R) signaling. TNFα promotes necroptosis, and its expression is facilitated by mitogen-activated protein (MAP) kinase-activated protein kinase 2 (MK2) but is inhibited by the RNA-binding protein tristetraprolin (TTP, encoded by the Zfp36 gene). We have stimulated murine macrophages from WT, MyD88-/-, Trif-/-, MyD88-/-Trif-/-, MK2-/-, and Zfp36-/- mice with graded doses of lipopolysaccharide (LPS) and various inhibitors to evaluate the role of various genes in Toll-like receptor 4 (TLR4)-induced necroptosis. Necrosome signaling, cytokine production, and cell death were evaluated by immunoblotting, ELISA, and cell death assays, respectively. We observed that during TLR4 signaling, necrosome activation is mediated through the adaptor proteins MyD88 and TRIF, and this is inhibited by MK2. In the absence of MK2-mediated necrosome activation, lipopolysaccharide-induced TNFα expression was drastically reduced, but MK2-deficient cells became highly sensitive to necroptosis even at low TNFα levels. In contrast, during tonic TLR4 signaling, WT cells did not undergo necroptosis, even when MK2 was disabled. Of note, necroptosis occurred only in the absence of TTP and was mediated by the expression of TNFα and activation of JUN N-terminal kinase (JNK). These results reveal that TTP plays an important role in inhibiting TNFα/JNK-induced necrosome signaling and resultant cytotoxicity.


Asunto(s)
Necroptosis , Transducción de Señal , Receptor Toll-Like 4/metabolismo , Tristetraprolina/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/deficiencia , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Clorometilcetonas de Aminoácidos/farmacología , Animales , Caspasa 8/química , Caspasa 8/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Necroptosis/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Tristetraprolina/deficiencia , Tristetraprolina/genética , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
6.
Proc Natl Acad Sci U S A ; 115(45): 11579-11584, 2018 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-30249666

RESUMEN

Adaptive natural killer (NK) cell memory represents a new frontier in immunology. Work over the last decade has discovered and confirmed the existence of NK cells with antigen-specific memories, which had previously been considered a unique property of T and B cells. These findings have shown that antigen-specific NK cells gain their specificity without the use of RAG proteins, representing a novel mechanism for generating antigen specificity, but the details of this mechanism have remained a mystery. We have discovered that members of the Ly49 family of surface receptors are critically involved in both the sensitization and the challenge phases of an NK cell memory response, as is antigen presentation from their binding partner, the class I MHC. Moreover, we demonstrate that the Ly49-interacting component of a presented antigen dictates the specificity of the NK cell memory response, implicating Ly49 receptors themselves in antigen-specific recognition. Finally, we demonstrate that adaptive NK cell memories can protect against an otherwise lethal melanoma without T cell or B cell support. These findings offer insight into the mechanism behind NK cell antigen specificity and demonstrate the clinical potential of this adaptive immune cell.


Asunto(s)
Dermatitis por Contacto/prevención & control , Memoria Inmunológica , Células Asesinas Naturales/inmunología , Melanoma Experimental/terapia , Subfamilia A de Receptores Similares a Lectina de Células NK/genética , Péptidos/inmunología , Inmunidad Adaptativa/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Presentación de Antígeno , Vacunas contra el Cáncer/administración & dosificación , Dermatitis por Contacto/genética , Dermatitis por Contacto/inmunología , Dermatitis por Contacto/patología , Dinitrofluorobenceno/administración & dosificación , Femenino , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/inmunología , Células Asesinas Naturales/citología , Células Asesinas Naturales/efectos de los fármacos , Masculino , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Subfamilia A de Receptores Similares a Lectina de Células NK/inmunología , Oxazoles/administración & dosificación , Péptidos/administración & dosificación , Péptidos/síntesis química , Vacunación
7.
J Biol Chem ; 293(30): 11913-11927, 2018 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-29899110

RESUMEN

Monocytes differentiate into macrophages, which deactivate invading pathogens. Macrophages can be resistant to cell death mechanisms in some situations, and the mechanisms involved are not clear. Here, using mouse immune cells, we investigated whether the differentiation of macrophages affects their susceptibility to cell death by the ripoptosome/necrosome pathways. We show that treatment of macrophages with a mimetic of second mitochondrial activator of caspases (SMAC) resulted in ripoptosome-driven cell death that specifically depended on tumor necrosis factor α (TNFα) expression and the receptor-interacting serine/threonine protein kinase 1 (RipK1)-RipK3-caspase-8 interaction in activated and cycling macrophages. Differentiation of macrophages increased the expression of pro-inflammatory cytokines but reduced RipK1-dependent cell death and the RipK3-caspase-8 interaction. The expression of the anti-apoptotic mediators, X-linked inhibitor of apoptosis protein (XIAP) and caspase-like apoptosis regulatory protein (cFLIPL), also increased in differentiated macrophages, which inhibited caspase activation. The resistance to cell death was abrogated in XIAP-deficient macrophages. However, even in the presence of increased XIAP expression, inhibition of the mitogen-activated protein kinase (MAPK) p38 and MAPK-activated protein kinase 2 (MK2) made differentiated macrophages susceptible to cell death. These results suggest that the p38/MK2 pathway overrides apoptosis inhibition by XIAP and that acquisition of resistance to cell death by increased expression of XIAP and cFLIPL may allow inflammatory macrophages to participate in pathogen control for a longer duration.


Asunto(s)
Inflamación/inmunología , Proteínas Inhibidoras de la Apoptosis/inmunología , Macrófagos/inmunología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología , Animales , Apoptosis , Diferenciación Celular , Células Cultivadas , Macrófagos/citología , Ratones Endogámicos C57BL
8.
Hum Mol Genet ; 26(4): 801-819, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28108555

RESUMEN

Spinal muscular atrophy (SMA) has long been solely considered a neurodegenerative disorder. However, recent work has highlighted defects in many other cell types that could contribute to disease aetiology. Interestingly, the immune system has never been extensively studied in SMA. Defects in lymphoid organs could exacerbate disease progression by neuroinflammation or immunodeficiency. Smn depletion led to severe alterations in the thymus and spleen of two different mouse models of SMA. The spleen from Smn depleted mice was dramatically smaller at a very young age and its histological architecture was marked by mislocalization of immune cells in the Smn2B/- model mice. In comparison, the thymus was relatively spared in gross morphology but showed many histological alterations including cortex thinning in both mouse models at symptomatic ages. Thymocyte development was also impaired as evidenced by abnormal population frequencies in the Smn2B/- thymus. Cytokine profiling revealed major changes in different tissues of both mouse models. Consistent with our observations, we found that survival motor neuron (Smn) protein levels were relatively high in lymphoid organs compared to skeletal muscle and spinal cord during postnatal development in wild type mice. Genetic introduction of one copy of the human SMN2 transgene was enough to rescue splenic and thymic defects in Smn2B/- mice. Thus, Smn is required for the normal development of lymphoid organs, and altered immune function may contribute to SMA disease pathogenesis.


Asunto(s)
Atrofia Muscular Espinal/inmunología , Proteína 1 para la Supervivencia de la Neurona Motora/inmunología , Timocitos/inmunología , Timo/inmunología , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/patología , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Timocitos/patología , Timo/patología
10.
J Immunol ; 196(7): 3097-108, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26895832

RESUMEN

Immune recognition of pathogen-associated ligands leads to assembly and activation of inflammasomes, resulting in the secretion of inflammatory cytokines IL-1ß and IL-18 and an inflammatory cell death called pyroptosis. Inflammasomes are important for protection against many pathogens, but their role during chronic infectious disease is poorly understood. Pseudomonas aeruginosa is an opportunistic pathogen that persists in the lungs of cystic fibrosis (CF) patients and may be responsible for the repeated episodes of pulmonary exacerbation characteristic of CF. P. aeruginosa is capable of inducing potent inflammasome activation during acute infection. We hypothesized that to persist within the host during chronic infection, P. aeruginosa must evade inflammasome activation, and pulmonary exacerbations may be the result of restoration of inflammasome activation. We therefore isolated P. aeruginosa from chronically infected CF patients during stable infection and exacerbation and evaluated the impact of these isolates on inflammasome activation in macrophages and neutrophils. P. aeruginosa isolates from CF patients failed to induce inflammasome activation, as measured by the secretion of IL-1ß and IL-18 and by pyroptotic cell death, during both stable infection and exacerbation. Inflammasome evasion likely was due to reduced expression of inflammasome ligands and reduced motility and was not observed in environmental isolates or isolates from acute, non-CF infection. These results reveal a novel mechanism of pathogen adaptation by P. aeruginosa to avoid detection by inflammasomes in CF patients and indicate that P. aeruginosa-activated inflammasomes are not involved in CF pulmonary exacerbations.


Asunto(s)
Fibrosis Quística/complicaciones , Inflamasomas/metabolismo , Infecciones por Pseudomonas/etiología , Infecciones por Pseudomonas/metabolismo , Animales , Caspasas/metabolismo , Línea Celular , Fibrosis Quística/inmunología , Citocinas/metabolismo , Progresión de la Enfermedad , Genes Bacterianos , Humanos , Ligandos , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Mutación , Neutrófilos/inmunología , Neutrófilos/metabolismo , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/inmunología , Pseudomonas aeruginosa/aislamiento & purificación , Esputo/microbiología , Sistemas de Secreción Tipo III/genética
11.
J Neural Transm (Vienna) ; 124(6): 721-738, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28477284

RESUMEN

Braak and Del Tredici have proposed that typical Parkinson disease (PD) has its origins in the olfactory bulb and gastrointestinal tract. However, the role of the olfactory system has insufficiently been explored in the pathogeneses of PD and Alzheimer disease (AD) in laboratory models. Here, we demonstrate applications of a new method to process mouse heads for microscopy by sectioning, mounting, and staining whole skulls ('holocranohistochemistry'). This technique permits the visualization of the olfactory system from the nasal cavity to mitral cells and dopamine-producing interneurons of glomeruli in the olfactory bulb. We applied this method to two specific goals: first, to visualize PD- and AD-linked gene expression in the olfactory system, where we detected abundant, endogenous α-synuclein and tau expression in the olfactory epithelium. Furthermore, we observed amyloid-ß plaques and proteinase-K-resistant α-synuclein species, respectively, in cranial nerve-I of APP- and human SNCA-over-expressing mice. The second application of the technique was to the modeling of gene-environment interactions in the nasal cavity of mice. We tracked the infection of a neurotropic respiratory-enteric-orphan virus from the nose pad into cranial nerves-I (and -V) and monitored the ensuing brain infection. Given its abundance in the olfactory epithelia, we questioned whether α-synuclein played a role in innate host defenses to modify the outcome of infections. Indeed, Snca-null mice were more likely to succumb to viral encephalitis versus their wild-type littermates. Moreover, using a bacterial sepsis model, Snca-null mice were less able to control infection after intravenous inoculation with Salmonella typhimurium. Together, holocranohistochemistry enabled new discoveries related to α-synuclein expression and its function in mice. Future studies will address: the role of Mapt and mutant SNCA alleles in infection paradigms; the contribution of xenobiotics in the initiation of idiopathic PD; and the safety to the host when systemically targeting α-synuclein by immunotherapy.


Asunto(s)
Encéfalo/metabolismo , Encéfalo/virología , Encefalitis Viral/virología , Mucosa Olfatoria/anatomía & histología , Mucosa Olfatoria/metabolismo , Infecciones por Reoviridae/virología , alfa-Sinucleína/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Modelos Animales de Enfermedad , Encefalitis Viral/inmunología , Encefalitis Viral/mortalidad , Encefalitis Viral/patología , Femenino , Cabeza , Humanos , Inmunohistoquímica , Masculino , Orthoreovirus Mamífero 3 , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Vías Nerviosas/anatomía & histología , Vías Nerviosas/diagnóstico por imagen , Vías Nerviosas/metabolismo , Vías Nerviosas/patología , Mucosa Olfatoria/patología , Neuronas Receptoras Olfatorias/metabolismo , Neuronas Receptoras Olfatorias/virología , Infecciones por Reoviridae/inmunología , Infecciones por Salmonella/inmunología , Infecciones por Salmonella/patología , Salmonella typhimurium , Conservación de Tejido/métodos , alfa-Sinucleína/genética
12.
Proc Natl Acad Sci U S A ; 111(31): E3206-13, 2014 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-25049377

RESUMEN

Myeloid cells play a critical role in perpetuating inflammation during various chronic diseases. Recently the death of macrophages through programmed necrosis (necroptosis) has emerged as an important mechanism in inflammation and pathology. We evaluated the mechanisms that lead to the induction of necrotic cell death in macrophages. Our results indicate that type I IFN (IFN-I) signaling is a predominant mechanism of necroptosis, because macrophages deficient in IFN-α receptor type I (IFNAR1) are highly resistant to necroptosis after stimulation with LPS, polyinosinic-polycytidylic acid, TNF-α, or IFN-ß in the presence of caspase inhibitors. IFN-I-induced necroptosis occurred through both mechanisms dependent on and independent of Toll/IL-1 receptor domain-containing adaptor inducing IFN-ß (TRIF) and led to persistent phosphorylation of receptor-interacting protein 3 (Rip3) kinase, which resulted in potent necroptosis. Although various IFN-regulatory factors (IRFs) facilitated the induction of necroptosis in response to IFN-ß, IRF-9-STAT1- or -STAT2-deficient macrophages were highly resistant to necroptosis. Our results indicate that IFN-ß-induced necroptosis of macrophages proceeds through tonic IFN-stimulated gene factor 3 (ISGF3) signaling, which leads to persistent expression of STAT1, STAT2, and IRF9. Induction of IFNAR1/Rip3-dependent necroptosis also resulted in potent inflammatory pathology in vivo. These results reveal how IFN-I mediates acute inflammation through macrophage necroptosis.


Asunto(s)
Apoptosis , Interferón Tipo I/metabolismo , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Transducción de Señal , Animales , Apoptosis/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Inflamación , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Necrosis , Oligopéptidos/farmacología , Poli I-C/farmacología , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/deficiencia , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología
13.
Immunol Cell Biol ; 94(4): 378-87, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26567886

RESUMEN

Salmonella enterica serovar Typhimurium (ST) is a virulent intracellular bacterium that conceals itself in the phagosomes of infected cells. Although CD8(+) T cells promote protection against various intracellular pathogens, the role of CD8(+) T cells against virulent ST has been unclear due to early fatality of susceptible (B6) mice. Herein, we generated MHC I-deficient mice on the resistant (129SvJ) and susceptible (Nramp1 transgenic B6) background to evaluate the role of CD8(+) T cells against virulent ST. Our results indicate that CD8(+) T cells have a critical protective role in host survival during infection with virulent ST. As antigen presentation and CD8(+) T-cell activation against phagosomal antigens are considered to operate through the cross-presentation pathway, we have evaluated CD8(+) T-cell response against ST in Batf3-deficient mice that lack CD8α dendritic cells (DCs). Using a recombinant of ST that expresses antigen (ST-OVA) mainly in the phagosomes of infected cells, we show that CD8(+) T-cell response is compromised throughout the duration of infection in Batf3-deficient mice. In contrast, when ST delivers antigen to the cytosol of infected cells (ST-OVA-C), CD8(+) T-cell response against the cytosolic antigen was compromised only in the short term in the absence of CD8α DCs, with wild-type and Batf3-deficient mice generating similar CD8(+) T-cell response in the long term. Thus, Batf3 has an important role in CD8(+) T-cell priming regardless of antigenic location; however, its role is redundant at later time intervals against cytosolic antigen.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Linfocitos T CD8-positivos/inmunología , Reactividad Cruzada , Fagosomas/metabolismo , Proteínas Represoras/metabolismo , Salmonella typhi/inmunología , Fiebre Tifoidea/inmunología , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Linfocitos T CD8-positivos/microbiología , Proteínas de Transporte de Catión/genética , Células Cultivadas , Citosol/metabolismo , Células Dendríticas/fisiología , Susceptibilidad a Enfermedades , Evasión Inmune , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Fagosomas/genética , Fagosomas/microbiología , Proteínas Represoras/genética
14.
J Immunol ; 192(12): 5671-8, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24799565

RESUMEN

It has recently been shown that programmed necrosis, necroptosis, may play a key role in the development of inflammation. Deciphering the regulation of this pathway within immune cells may therefore have implications in pathology associated with inflammatory diseases. We show that treatment of macrophages with the pan caspase inhibitor (zVAD-FMK) results in both increased phosphorylation and decreased cleavage of receptor interacting protein kinase-1 (Rip1), leading to necroptosis that is dependent on autocrine TNF signaling. Stimulation of cells with TLR agonists such as LPS in the presence of zVAD-FMK also induced Rip1-phosphorylation via a TNFR-independent mechanism. Further examination of Rip1 expression under these stimulatory conditions revealed a regulatory cleavage of Rip1 in macrophages that is not apparently attributable to caspase-8. Instead, we provide novel evidence that cysteine family cathepsins, which are highly abundant in myeloid cells, can also cleave Rip1 kinase. Using small interfering RNA knockdown, specific cathepsin inhibitors, and cell-free cleavage assays, we demonstrate that cysteine cathepsins B and S can directly cleave Rip1. Finally, we demonstrate that only through combined inhibition of cathepsins and caspase-8 could a potent induction of macrophage necroptosis be achieved. These data reveal a novel mechanism of regulation of necroptosis by cathepsins within macrophage cells.


Asunto(s)
Catepsina B/inmunología , Catepsinas/inmunología , Macrófagos/inmunología , Proteolisis , Proteína Serina-Treonina Quinasas de Interacción con Receptores/inmunología , Clorometilcetonas de Aminoácidos/farmacología , Animales , Caspasa 8/genética , Caspasa 8/inmunología , Inhibidores de Caspasas/farmacología , Catepsina B/antagonistas & inhibidores , Catepsina B/genética , Catepsinas/antagonistas & inhibidores , Catepsinas/genética , Lipopolisacáridos/farmacología , Macrófagos/citología , Ratones , Ratones Noqueados , Fosforilación/efectos de los fármacos , Fosforilación/genética , Fosforilación/inmunología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Receptores Toll-Like/agonistas , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología
15.
Cell Immunol ; 293(2): 122-5, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25596474

RESUMEN

Many soluble cytokine receptors inhibit cytokine bioactivity, while others prolong ligand activity. The biological role of an endogenous soluble form of IL-7Rα, or its therapeutic effects on CD8(+) T-cells are unknown. We demonstrate that recombinant IL-7Rα-Fc, when pre-incubated with IL-7, enhances IL-7-induced CD8(+) T-cell proliferation and viability of human or murine CD8(+) T-cells. Receptor blocking experiments confirmed IL-7-specific activity. These data demonstrate that exogenous soluble IL-7Rα significantly enhances CD8(+) T-cell responses to IL-7 in vitro and paves the way for future research to determine its therapeutic potential to restore impaired CD8(+) T-cell function in disease.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Proliferación Celular/efectos de los fármacos , Interleucina-7/inmunología , Receptores de Interleucina-7/inmunología , Animales , Humanos , Ratones , Ratones Endogámicos C57BL
16.
J Immunol ; 190(3): 1066-75, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23277488

RESUMEN

CD8(+) T cells undergo rapid expansion during infection with intracellular pathogens, which is followed by swift and massive culling of primed CD8(+) T cells. The mechanisms that govern the massive contraction and maintenance of primed CD8(+) T cells are not clear. We show in this study that the transcription factor, FoxO3a, does not influence Ag presentation and the consequent expansion of CD8(+) T cell response during Listeria monocytogenes infection, but plays a key role in the maintenance of memory CD8(+) T cells. The effector function of primed CD8(+) T cells as revealed by cytokine secretion and CD107a degranulation was not influenced by inactivation of FoxO3a. Interestingly, FoxO3a-deficient CD8(+) T cells displayed reduced expression of proapoptotic molecules BIM and PUMA during the various phases of response, and underwent reduced apoptosis in comparison with wild-type cells. A higher number of memory precursor effector cells and memory subsets was detectable in FoxO3a-deficient mice compared with wild-type mice. Furthermore, FoxO3a-deficient memory CD8(+) T cells upon transfer into normal or RAG1-deficient mice displayed enhanced survival. These results suggest that FoxO3a acts in a cell-intrinsic manner to regulate the survival of primed CD8(+) T cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Factores de Transcripción Forkhead/inmunología , Memoria Inmunológica/inmunología , Listeriosis/inmunología , Activación de Linfocitos/inmunología , Subgrupos Linfocitarios/inmunología , Animales , Presentación de Antígeno , Antígenos Bacterianos/inmunología , Apoptosis/inmunología , Proteínas Reguladoras de la Apoptosis/biosíntesis , Proteínas Reguladoras de la Apoptosis/genética , Proteína 11 Similar a Bcl2 , Linfocitos T CD8-positivos/metabolismo , Citocinas/sangre , Citotoxicidad Inmunológica , Femenino , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/deficiencia , Proteínas de Homeodominio/genética , Selectina L/biosíntesis , Selectina L/genética , Listeria monocytogenes/inmunología , Listeriosis/sangre , Subgrupos Linfocitarios/metabolismo , Linfocinas/metabolismo , Proteínas de Membrana de los Lisosomas/inmunología , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/genética , Ovalbúmina/inmunología , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/genética , Receptores de Interleucina-7/biosíntesis , Receptores de Interleucina-7/genética , Proteínas Supresoras de Tumor/biosíntesis , Proteínas Supresoras de Tumor/genética
17.
J Immunol ; 187(3): 1192-200, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21715683

RESUMEN

After vaccination, memory CD8(+) T cells migrate to different organs to mediate immune surveillance. In most nonlymphoid organs, following an infection, CD8(+) T cells differentiate to become long-lived effector-memory cells, thereby providing long-term protection against a secondary infection. In this study, we demonstrated that Ag-specific CD8(+) T cells that migrate to the mouse brain following a systemic Listeria infection do not display markers reminiscent of long-term memory cells. In contrast to spleen and other nonlymphoid organs, none of the CD8(+) T cells in the brain reverted to a memory phenotype, and all of the cells were gradually eliminated. These nonmemory phenotype CD8(+) T cells were found primarily within the choroid plexus, as well as in the cerebrospinal fluid-filled spaces. Entry of these CD8(+) T cells into the brain was governed primarily by CD49d/VCAM-1, with the majority of entry occurring in the first week postinfection. When CD8(+) T cells were injected directly into the brain parenchyma, cells that remained in the brain retained a highly activated (CD69(hi)) phenotype and were gradually lost, whereas those that migrated out to the spleen were CD69(low) and persisted long-term. These results revealed a mechanism of time-bound immune surveillance to the brain by CD8(+) T cells that do not reside in the parenchyma.


Asunto(s)
Encéfalo/inmunología , Linfocitos T CD8-positivos/inmunología , Vigilancia Inmunológica , Listeriosis/líquido cefalorraquídeo , Listeriosis/inmunología , Activación de Linfocitos/inmunología , Animales , Encéfalo/microbiología , Encéfalo/patología , Linfocitos T CD8-positivos/microbiología , Linfocitos T CD8-positivos/trasplante , Movimiento Celular/inmunología , Supervivencia Celular/inmunología , Células Cultivadas , Epítopos de Linfocito T/líquido cefalorraquídeo , Epítopos de Linfocito T/inmunología , Femenino , Memoria Inmunológica , Inmunofenotipificación , Listeria monocytogenes/inmunología , Listeriosis/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
18.
Cell Mol Immunol ; 20(10): 1140-1155, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37553427

RESUMEN

Natural killer (NK) cells are predominant innate lymphocytes that initiate the early immune response during infection. NK cells undergo a metabolic switch to fuel augmented proliferation and activation following infection. Tumor necrosis factor-alpha (TNFα) is a well-known inflammatory cytokine that enhances NK cell function; however, the mechanism underlying NK cell proliferation in response to TNFα is not well established. Here, we demonstrated that upon infection/inflammation, NK cells upregulate the expression of TNF receptor 2 (TNFR2), which is associated with increased proliferation, metabolic activity, and effector function. Notably, IL-18 can induce TNFR2 expression in NK cells, augmenting their sensitivity toward TNFα. Mechanistically, TNFα-TNFR2 signaling upregulates the expression of CD25 (IL-2Rα) and nutrient transporters in NK cells, leading to a metabolic switch toward aerobic glycolysis. Transcriptomic analysis revealed significantly reduced expression levels of genes involved in cellular metabolism and proliferation in NK cells from TNFR2 KO mice. Accordingly, our data affirmed that genetic ablation of TNFR2 curtails CD25 upregulation and TNFα-induced glycolysis, leading to impaired NK cell proliferation and antiviral function during MCMV infection in vivo. Collectively, our results delineate the crucial role of the TNFα-TNFR2 axis in NK cell proliferation, glycolysis, and effector function.


Asunto(s)
Receptores Tipo II del Factor de Necrosis Tumoral , Factor de Necrosis Tumoral alfa , Ratones , Animales , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Células Asesinas Naturales , Citocinas/metabolismo , Proliferación Celular
19.
Am J Reprod Immunol ; 88(4): e13599, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35851978

RESUMEN

PROBLEM: Salmonella enterica serovar Typhimurium (S.Tm) infection in Nramp1+/+ mice during pregnancy can lead to profound bacterial growth in the feto-placental unit and adverse pregnancy outcomes, including fetal loss, maternal illness and death. The kinetics and mechanisms by which S.Tm gains entry within individual feto-placental unit, and disseminates through tissues leading to placental resorption and fetal demise remain unclear. METHOD OF STUDY: Mice were systemically infected with S.Tm. Bacterial burden within spleen and individual placentas, and placental/fetal resorptions were quantified. Flow cytometric analysis of immune cell types in the spleen and individual placentas was performed. Cytokine expression in maternal serum was determined through cytometric bead array. RESULTS: Systemic infection with S.Tm resulted in preferential bacterial proliferation in placentas compared to the spleen in Nramp1+/+ mice. At 24 h post-infection, the mean infection rate of individual placentas per mouse was ∼50%, increasing to >75% by 72 h post-infection, suggesting that initial infection in few sites progresses to rapid spread of infection through the uterine milieu. This correlated with a steady increase in placental/fetal resorption rates. Placental infection was associated with local increased neutrophil percentages, whereas numbers and percentages in the spleen remained unchanged, suggesting dichotomous modulation of inflammation between the systemic compartment and the feto-maternal interface. Reduced survival rates of pregnant mice during infection correlated with decreased serum IFN-γ but increased IL-10 levels relative to non-pregnant controls. CONCLUSION: Pregnancy compromises host resistance conferred by Nramp1 against S.Tm through compartment-specific regulation of maternal and placental cellular responses, and modulation of systemic cytokine expression.


Asunto(s)
Interleucina-10 , Infecciones por Salmonella , Animales , Proteínas de Transporte de Catión , Citocinas , Femenino , Inmunidad , Ratones , Placenta , Embarazo , Salmonella typhimurium , Serogrupo
20.
Cell Death Differ ; 29(3): 585-599, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34588632

RESUMEN

Mutations in susceptibility alleles correlate with gut-inflammatory diseases, such as Crohn's disease; however, this does not often impact the disease progression indicating the existence of compensatory genes. We show that a reduction in Foxo3a expression in IL-10-deficient mice results in a spontaneous and aggressive Crohn's- like disease with 100% penetrance, which is rescued by deletion of myeloid cells, T cells and inhibition of mTORC1. In Foxo3a-/- IL-10-/- mice, there is poor cell death of myeloid cells in the gut, leading to increased accumulation of myeloid and T cells in the gut. Myeloid cells express high levels of inflammatory cytokines, and regulatory T cells are dysfunctional despite increased abundance. Foxo3a signaling represses the transcription of glutaminase (GLS/GLS2) to prevent over-consumption of glutamine by activated T cells and its conversion to glutamate that contributes to the TCA cycle and mTORC1 activation. Finally, we show that Foxo3a restricts the abundance of colitogenic microbiota in IL-10-deficient mice. Thus, by suppressing glutaminolysis in activated T cells Foxo3a mediates a critical checkpoint that prevents the development of fulminant gut inflammatory disease.


Asunto(s)
Colitis , Proteína Forkhead Box O3/metabolismo , Interleucina-10 , Animales , Colitis/genética , Colitis/prevención & control , Inflamación , Interleucina-10/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Ratones , Linfocitos T
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA