Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.187
Filtrar
1.
Cell ; 186(11): 2288-2312, 2023 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-37236155

RESUMEN

Inflammasomes are critical sentinels of the innate immune system that respond to threats to the host through recognition of distinct molecules, known as pathogen- or damage-associated molecular patterns (PAMPs/DAMPs), or disruptions of cellular homeostasis, referred to as homeostasis-altering molecular processes (HAMPs) or effector-triggered immunity (ETI). Several distinct proteins nucleate inflammasomes, including NLRP1, CARD8, NLRP3, NLRP6, NLRC4/NAIP, AIM2, pyrin, and caspases-4/-5/-11. This diverse array of sensors strengthens the inflammasome response through redundancy and plasticity. Here, we present an overview of these pathways, outlining the mechanisms of inflammasome formation, subcellular regulation, and pyroptosis, and discuss the wide-reaching effects of inflammasomes in human disease.


Asunto(s)
Inflamasomas , Humanos , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Adaptadoras de Señalización CARD/metabolismo , Caspasas/metabolismo , Muerte Celular , Inflamasomas/metabolismo , Proteínas de Neoplasias/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Piroptosis
2.
Annu Rev Immunol ; 33: 49-77, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25493334

RESUMEN

Induction, production, and release of proinflammatory cytokines are essential steps to establish an effective host defense. Cytokines of the interleukin-1 (IL-1) family induce inflammation and regulate T lymphocyte responses while also displaying homeostatic and metabolic activities. With the exception of the IL-1 receptor antagonist, all IL-1 family cytokines lack a signal peptide and require proteolytic processing into an active molecule. One such unique protease is caspase-1, which is activated by protein platforms called the inflammasomes. However, increasing evidence suggests that inflammasomes and caspase-1 are not the only mechanism for processing IL-1 cytokines. IL-1 cytokines are often released as precursors and require extracellular processing for activity. Here we review the inflammasome-independent enzymatic processes that are able to activate IL-1 cytokines, paying special attention to neutrophil-derived serine proteases, which subsequently induce inflammation and modulate host defense. The inflammasome-independent processing of IL-1 cytokines has important consequences for understanding inflammatory diseases, and it impacts the design of IL-1-based modulatory therapies.


Asunto(s)
Citocinas/metabolismo , Inflamasomas/metabolismo , Interleucina-1/metabolismo , Animales , Susceptibilidad a Enfermedades , Humanos , Inflamación/metabolismo , Mediadores de Inflamación/metabolismo
3.
Immunity ; 57(4): 674-699, 2024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38599165

RESUMEN

Nucleotide-binding oligomerization domain (NOD)-like receptors, also known as nucleotide-binding leucine-rich repeat receptors (NLRs), are a family of cytosolic pattern recognition receptors that detect a wide variety of pathogenic and sterile triggers. Activation of specific NLRs initiates pro- or anti-inflammatory signaling cascades and the formation of inflammasomes-multi-protein complexes that induce caspase-1 activation to drive inflammatory cytokine maturation and lytic cell death, pyroptosis. Certain NLRs and inflammasomes act as integral components of larger cell death complexes-PANoptosomes-driving another form of lytic cell death, PANoptosis. Here, we review the current understanding of the evolution, structure, and function of NLRs in health and disease. We discuss the concept of NLR networks and their roles in driving cell death and immunity. An improved mechanistic understanding of NLRs may provide therapeutic strategies applicable across infectious and inflammatory diseases and in cancer.


Asunto(s)
Inflamasomas , Receptores de Reconocimiento de Patrones , Inflamasomas/metabolismo , Piroptosis , Inmunidad Innata , Nucleótidos
4.
Immunity ; 56(8): 1825-1843.e6, 2023 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-37451265

RESUMEN

Glioblastoma (GBM), a highly lethal brain cancer, is notorious for immunosuppression, but the mechanisms remain unclear. Here, we documented a temporospatial patterning of tumor-associated myeloid cells (TAMs) corresponding to vascular changes during GBM progression. As tumor vessels transitioned from the initial dense regular network to later scant and engorged vasculature, TAMs shifted away from perivascular regions and trafficked to vascular-poor areas. This process was heavily influenced by the immunocompetence state of the host. Utilizing a sensitive fluorescent UnaG reporter to track tumor hypoxia, coupled with single-cell transcriptomics, we revealed that hypoxic niches attracted and sequestered TAMs and cytotoxic T lymphocytes (CTLs), where they were reprogrammed toward an immunosuppressive state. Mechanistically, we identified chemokine CCL8 and cytokine IL-1ß as two hypoxic-niche factors critical for TAM trafficking and co-evolution of hypoxic zones into pseudopalisading patterns. Therefore, perturbation of TAM patterning in hypoxic zones may improve tumor control.


Asunto(s)
Glioblastoma , Linfocitos T Citotóxicos , Humanos , Macrófagos Asociados a Tumores , Macrófagos , Terapia de Inmunosupresión , Glioblastoma/patología , Microambiente Tumoral
5.
Cell ; 171(5): 1110-1124.e18, 2017 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-29033128

RESUMEN

Detection of cytosolic DNA constitutes a central event in the context of numerous infectious and sterile inflammatory conditions. Recent studies have uncovered a bipartite mode of cytosolic DNA recognition, in which the cGAS-STING axis triggers antiviral immunity, whereas AIM2 triggers inflammasome activation. Here, we show that AIM2 is dispensable for DNA-mediated inflammasome activation in human myeloid cells. Instead, detection of cytosolic DNA by the cGAS-STING axis induces a cell death program initiating potassium efflux upstream of NLRP3. Forward genetics identified regulators of lysosomal trafficking to modulate this cell death program, and subsequent studies revealed that activated STING traffics to the lysosome, where it triggers membrane permeabilization and thus lysosomal cell death (LCD). Importantly, the cGAS-STING-NLRP3 pathway constitutes the default inflammasome response during viral and bacterial infections in human myeloid cells. We conclude that targeting the cGAS-STING-LCD-NLRP3 pathway will ameliorate pathology in inflammatory conditions that are associated with cytosolic DNA sensing.


Asunto(s)
Muerte Celular , Inflamasomas/metabolismo , Monocitos/citología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , ADN/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Monocitos/metabolismo , Transducción de Señal
6.
Cell ; 171(5): 1057-1071.e11, 2017 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-29033131

RESUMEN

Type I interferon restrains interleukin-1ß (IL-1ß)-driven inflammation in macrophages by upregulating cholesterol-25-hydroxylase (Ch25h) and repressing SREBP transcription factors. However, the molecular links between lipid metabolism and IL-1ß production remain obscure. Here, we demonstrate that production of 25-hydroxycholesterol (25-HC) by macrophages is required to prevent inflammasome activation by the DNA sensor protein absent in melanoma 2 (AIM2). We find that in response to bacterial infection or lipopolysaccharide (LPS) stimulation, macrophages upregulate Ch25h to maintain repression of SREBP2 activation and cholesterol synthesis. Increasing macrophage cholesterol content is sufficient to trigger IL-1ß release in a crystal-independent but AIM2-dependent manner. Ch25h deficiency results in cholesterol-dependent reduced mitochondrial respiratory capacity and release of mitochondrial DNA into the cytosol. AIM2 deficiency rescues the increased inflammasome activity observed in Ch25h-/-. Therefore, activated macrophages utilize 25-HC in an anti-inflammatory circuit that maintains mitochondrial integrity and prevents spurious AIM2 inflammasome activation.


Asunto(s)
Colesterol/metabolismo , Inflamasomas/metabolismo , Macrófagos/metabolismo , Animales , Colesterol/biosíntesis , ADN Mitocondrial/metabolismo , Proteínas de Unión al ADN/metabolismo , Humanos , Hidroxicolesteroles/metabolismo , Inflamasomas/inmunología , Inflamación/inmunología , Inflamación/patología , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Listeria monocytogenes/fisiología , Listeriosis/inmunología , Macrófagos/citología , Macrófagos/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Mitocondrias/metabolismo , Oxiesteroles/metabolismo
7.
Cell ; 168(3): 503-516.e12, 2017 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-28129542

RESUMEN

Sickness-induced anorexia is a conserved behavior induced during infections. Here, we report that an intestinal pathogen, Salmonella Typhimurium, inhibits anorexia by manipulating the gut-brain axis. Inhibition of inflammasome activation by the S. Typhimurium effector, SlrP, prevented anorexia caused by IL-1ß-mediated signaling to the hypothalamus via the vagus nerve. Rather than compromising host defenses, pathogen-mediated inhibition of anorexia increased host survival. SlrP-mediated inhibition of anorexia prevented invasion and systemic infection by wild-type S. Typhimurium, reducing virulence while increasing transmission to new hosts, suggesting that there are trade-offs between transmission and virulence. These results clarify the complex and contextual role of anorexia in host-pathogen interactions and suggest that microbes have evolved mechanisms to modulate sickness-induced behaviors to promote health of their host and their transmission at the expense of virulence.


Asunto(s)
Anorexia/microbiología , Infecciones por Salmonella/microbiología , Infecciones por Salmonella/transmisión , Salmonella typhimurium/patogenicidad , Animales , Proteínas Bacterianas/metabolismo , Interacciones Huésped-Patógeno , Humanos , Inflamasomas/inmunología , Interleucina-1beta/inmunología , Ratones , Ratones Endogámicos C57BL , Vías Nerviosas , Infecciones por Salmonella/inmunología , Salmonella typhimurium/fisiología , Organismos Libres de Patógenos Específicos , Virulencia
8.
Immunity ; 55(9): 1645-1662.e7, 2022 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-35882236

RESUMEN

Healthy skin maintains a diverse microbiome and a potent immune system to fight off infections. Here, we discovered that the epithelial-cell-derived antimicrobial peptides defensins activated orphan G-protein-coupled receptors (GPCRs) Mrgpra2a/b on neutrophils. This signaling axis was required for effective neutrophil-mediated skin immunity and microbiome homeostasis. We generated mutant mouse lines lacking the entire Defensin (Def) gene cluster in keratinocytes or Mrgpra2a/b. Def and Mrgpra2 mutant animals both exhibited skin dysbiosis, with reduced microbial diversity and expansion of Staphylococcus species. Defensins and Mrgpra2 were critical for combating S. aureus infections and the formation of neutrophil abscesses, a hallmark of antibacterial immunity. Activation of Mrgpra2 by defensin triggered neutrophil release of IL-1ß and CXCL2 which are vital for proper amplification and propagation of the antibacterial immune response. This study demonstrated the importance of epithelial-neutrophil signaling via the defensin-Mrgpra2 axis in maintaining healthy skin ecology and promoting antibacterial host defense.


Asunto(s)
Infecciones Bacterianas , Neutrófilos , Receptores Acoplados a Proteínas G , Animales , Ratones , Antibacterianos , Proteínas Portadoras , Defensinas/genética , Disbiosis , Queratinocitos , Receptores Acoplados a Proteínas G/metabolismo , Staphylococcus aureus
9.
Mol Cell ; 83(11): 1872-1886.e5, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37172591

RESUMEN

Deregulated inflammation is a critical feature driving the progression of tumors harboring mutations in the liver kinase B1 (LKB1), yet the mechanisms linking LKB1 mutations to deregulated inflammation remain undefined. Here, we identify deregulated signaling by CREB-regulated transcription coactivator 2 (CRTC2) as an epigenetic driver of inflammatory potential downstream of LKB1 loss. We demonstrate that LKB1 mutations sensitize both transformed and non-transformed cells to diverse inflammatory stimuli, promoting heightened cytokine and chemokine production. LKB1 loss triggers elevated CRTC2-CREB signaling downstream of the salt-inducible kinases (SIKs), increasing inflammatory gene expression in LKB1-deficient cells. Mechanistically, CRTC2 cooperates with the histone acetyltransferases CBP/p300 to deposit histone acetylation marks associated with active transcription (i.e., H3K27ac) at inflammatory gene loci, promoting cytokine expression. Together, our data reveal a previously undefined anti-inflammatory program, regulated by LKB1 and reinforced through CRTC2-dependent histone modification signaling, that links metabolic and epigenetic states to cell-intrinsic inflammatory potential.


Asunto(s)
Histonas , Proteínas Serina-Treonina Quinasas , Humanos , Histonas/genética , Histonas/metabolismo , Acetilación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Citocinas/metabolismo , Inflamación/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
10.
Immunity ; 54(7): 1463-1477.e11, 2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34115964

RESUMEN

Acute respiratory distress syndrome (ARDS), an inflammatory condition with high mortality rates, is common in severe COVID-19, whose risk is reduced by metformin rather than other anti-diabetic medications. Detecting of inflammasome assembly in post-mortem COVID-19 lungs, we asked whether and how metformin inhibits inflammasome activation while exerting its anti-inflammatory effect. We show that metformin inhibited NLRP3 inflammasome activation and interleukin (IL)-1ß production in cultured and alveolar macrophages along with inflammasome-independent IL-6 secretion, thus attenuating lipopolysaccharide (LPS)- and SARS-CoV-2-induced ARDS. By targeting electron transport chain complex 1 and independently of AMP-activated protein kinase (AMPK) or NF-κB, metformin blocked LPS-induced and ATP-dependent mitochondrial (mt) DNA synthesis and generation of oxidized mtDNA, an NLRP3 ligand. Myeloid-specific ablation of LPS-induced cytidine monophosphate kinase 2 (CMPK2), which is rate limiting for mtDNA synthesis, reduced ARDS severity without a direct effect on IL-6. Thus, inhibition of ATP and mtDNA synthesis is sufficient for ARDS amelioration.


Asunto(s)
Adenosina Trifosfato/metabolismo , ADN Mitocondrial/biosíntesis , Inflamasomas/efectos de los fármacos , Metformina/farmacología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neumonía/prevención & control , Animales , COVID-19/metabolismo , COVID-19/prevención & control , Citocinas/genética , Citocinas/metabolismo , ADN Mitocondrial/metabolismo , Humanos , Inflamasomas/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Lipopolisacáridos/toxicidad , Metformina/uso terapéutico , Ratones , Nucleósido-Fosfato Quinasa/metabolismo , Neumonía/metabolismo , Síndrome de Dificultad Respiratoria/inducido químicamente , Síndrome de Dificultad Respiratoria/prevención & control , SARS-CoV-2/patogenicidad
11.
Immunity ; 52(2): 342-356.e6, 2020 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-32023490

RESUMEN

Interleukin-17A (IL-17A) is a major mediator of tissue inflammation in many autoimmune diseases. Anti-IL-17A is an effective treatment for psoriasis and is showing promise in clinical trials in multiple sclerosis. In this study, we find that IL-17A-defective mice or mice treated with anti-IL-17A at induction of experimental autoimmune encephalomyelitis (EAE) are resistant to disease and have defective priming of IL-17-secreting γδ T (γδT17) cells and Th17 cells. However, T cells from Il17a-/- mice induce EAE in wild-type mice following in vitro culture with autoantigen, IL-1ß, and IL-23. Furthermore, treatment with IL-1ß or IL-17A at induction of EAE restores disease in Il17a-/- mice. Importantly, mobilization of IL-1ß-producing neutrophils and inflammatory monocytes and activation of γδT17 cells is reduced in Il17a-/- mice. Our findings demonstrate that a key function of IL-17A in central nervous system (CNS) autoimmunity is to recruit IL-1ß-secreting myeloid cells that prime pathogenic γδT17 and Th17 cells.


Asunto(s)
Autoinmunidad/inmunología , Interleucina-17/inmunología , Interleucina-1beta/metabolismo , Linfocitos Intraepiteliales/inmunología , Células Mieloides/inmunología , Células Th17/inmunología , Animales , Autoantígenos/inmunología , Autoinmunidad/genética , Sistema Nervioso Central/inmunología , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Interleucina-17/antagonistas & inhibidores , Interleucina-17/deficiencia , Interleucina-17/metabolismo , Interleucina-1beta/inmunología , Interleucina-23/inmunología , Interleucina-23/metabolismo , Linfocitos Intraepiteliales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/inmunología , Monocitos/metabolismo , Células Mieloides/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Células Th17/metabolismo
12.
Immunity ; 50(4): 1099-1114.e10, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30876876

RESUMEN

Inflammatory bowel disease is a chronic, relapsing condition with two subtypes, Crohn's disease (CD) and ulcerative colitis (UC). Genome-wide association studies (GWASs) in UC implicate a FCGR2A variant that alters the binding affinity of the antibody receptor it encodes, FcγRIIA, for immunoglobulin G (IgG). Here, we aimed to understand the mechanisms whereby changes in FcγRIIA affinity would affect inflammation in an IgA-dominated organ. We found a profound induction of anti-commensal IgG and a concomitant increase in activating FcγR signaling in the colonic mucosa of UC patients. Commensal-IgG immune complexes engaged gut-resident FcγR-expressing macrophages, inducing NLRP3- and reactive-oxygen-species-dependent production of interleukin-1ß (IL-1ß) and neutrophil-recruiting chemokines. These responses were modulated by the FCGR2A genotype. In vivo manipulation of macrophage FcγR signal strength in a mouse model of UC determined the magnitude of intestinal inflammation and IL-1ß-dependent type 17 immunity. The identification of an important contribution of IgG-FcγR-dependent inflammation to UC has therapeutic implications.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Colitis Ulcerosa/inmunología , Microbioma Gastrointestinal/inmunología , Inmunoglobulina G/inmunología , Interleucina-1beta/inmunología , Células Th17/inmunología , Animales , Colitis/inducido químicamente , Colitis/inmunología , Colitis/microbiología , Colitis/patología , Colitis Ulcerosa/microbiología , Colitis Ulcerosa/patología , Sulfato de Dextran/toxicidad , Regulación de la Expresión Génica , Genotipo , Humanos , Inflamación , Interleucina-8/biosíntesis , Interleucina-8/genética , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Macrófagos/inmunología , Ratones , Fagocitos/inmunología , ARN Mensajero/biosíntesis , Especies Reactivas de Oxígeno , Receptores de IgG/biosíntesis , Receptores de IgG/genética , Receptores de IgG/inmunología
13.
Immunity ; 48(1): 75-90.e6, 2018 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-29343442

RESUMEN

The molecular basis of signal-dependent transcriptional activation has been extensively studied in macrophage polarization, but our understanding remains limited regarding the molecular determinants of repression. Here we show that IL-4-activated STAT6 transcription factor is required for the direct transcriptional repression of a large number of genes during in vitro and in vivo alternative macrophage polarization. Repression results in decreased lineage-determining transcription factor, p300, and RNA polymerase II binding followed by reduced enhancer RNA expression, H3K27 acetylation, and chromatin accessibility. The repressor function of STAT6 is HDAC3 dependent on a subset of IL-4-repressed genes. In addition, STAT6-repressed enhancers show extensive overlap with the NF-κB p65 cistrome and exhibit decreased responsiveness to lipopolysaccharide after IL-4 stimulus on a subset of genes. As a consequence, macrophages exhibit diminished inflammasome activation, decreased IL-1ß production, and pyroptosis. Thus, the IL-4-STAT6 signaling pathway establishes an alternative polarization-specific epigenenomic signature resulting in dampened macrophage responsiveness to inflammatory stimuli.


Asunto(s)
Interleucina-4/metabolismo , Macrófagos/metabolismo , Factor de Transcripción STAT6/metabolismo , Animales , Western Blotting , Línea Celular , Elementos de Facilitación Genéticos , Citometría de Flujo , Regulación de la Expresión Génica , Inflamasomas/metabolismo , Citometría de Barrido por Láser , Lipopolisacáridos/farmacología , Macrófagos/fisiología , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa , Piroptosis/genética , Transducción de Señal/genética , Transducción de Señal/fisiología
14.
Proc Natl Acad Sci U S A ; 121(8): e2312621121, 2024 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-38346191

RESUMEN

One of the hallmarks of type 1 but also type 2 diabetes is pancreatic islet inflammation, associated with altered pancreatic islet function and structure, if unresolved. IL-1ß is a proinflammatory cytokine which detrimentally affects ß-cell function. In the course of diabetes, complement components, including the central complement protein C3, are deregulated. Previously, we reported high C3 expression in human pancreatic islets, with upregulation after IL-1ß treatment. In the current investigation, using primary human and rodent material and CRISPR/Cas9 gene-edited ß-cells deficient in C3, or producing only cytosolic C3 from a noncanonical in-frame start codon, we report a protective effect of C3 against IL-1ß-induced ß-cell death, that is attributed to the cytosolic fraction of C3. Further investigation revealed that intracellular C3 alleviates IL-1ß-induced ß-cell death, by interaction with and inhibition of Fyn-related kinase (FRK), which is involved in the response of ß-cells to cytokines. Furthermore, these data were supported by increased ß-cell death in vivo in a ß-cell-specific C3 knockout mouse. Our data indicate that a functional, cytoprotective association exists between FRK and cytosolic C3.


Asunto(s)
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Islotes Pancreáticos , Ratones , Animales , Humanos , Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Muerte Celular , Citocinas/metabolismo , Ratones Noqueados
15.
Immunity ; 47(3): 510-523.e4, 2017 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-28930661

RESUMEN

Within the interleukin 1 (IL-1) cytokine family, IL-1 receptor accessory protein (IL-1RAcP) is the co-receptor for eight receptor-cytokine pairs, including those involving cytokines IL-1ß and IL-33. Unlike IL-1ß, IL-33 does not have a signaling complex that includes both its cognate receptor, ST2, and the shared co-receptor IL-1RAcP, which we now present here. Although the IL-1ß and IL-33 complexes shared structural features and engaged identical molecular surfaces of IL-1RAcP, these cytokines had starkly different strategies for co-receptor engagement and signal activation. Our data suggest that IL-1ß binds to IL-1RI to properly present the cytokine to IL-1RAcP, whereas IL-33 binds to ST2 in order to conformationally constrain the cognate receptor in an IL-1RAcP-receptive state. These findings indicate that members of the IL-1 family of cytokines use distinct molecular mechanisms to signal through their shared co-receptor, and they provide the foundation from which to design new therapies to target IL-33 signaling.


Asunto(s)
Interleucina-1/metabolismo , Receptores de Interleucina-1/metabolismo , Transducción de Señal , Animales , Sitios de Unión , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Interleucina-1/química , Proteína 1 Similar al Receptor de Interleucina-1/química , Proteína 1 Similar al Receptor de Interleucina-1/genética , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Interleucina-33/química , Interleucina-33/metabolismo , Ratones , Modelos Moleculares , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Mutación , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Receptores de Interleucina-1/química , Receptores de Interleucina-1/genética
16.
Immunity ; 47(5): 959-973.e9, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29150241

RESUMEN

Aortic aneurysms are life-threatening conditions with effective treatments mainly limited to emergency surgery or trans-arterial endovascular stent grafts, thus calling for the identification of specific molecular targets. Genetic studies have highlighted controversial roles of transforming growth factor ß (TGF-ß) signaling in aneurysm development. Here, we report on aneurysms developing in adult mice after smooth muscle cell (SMC)-specific inactivation of Smad4, an intracellular transducer of TGF-ß. The results revealed that Smad4 inhibition activated interleukin-1ß (IL-1ß) in SMCs. This danger signal later recruited innate immunity in the adventitia through chemokine (C-C motif) ligand 2 (CCL2) and modified the mechanical properties of the aortic wall, thus favoring vessel dilation. SMC-specific Smad4 deletion in Il1r1- or Ccr2-null mice resulted in milder aortic pathology. A chronic treatment with anti-IL-1ß antibody effectively hampered aneurysm development. These findings identify a mechanistic target for controlling the progression of aneurysms with compromised TGF-ß signaling, such as those driven by SMAD4 mutations.


Asunto(s)
Aneurisma de la Aorta/prevención & control , Interleucina-1beta/antagonistas & inhibidores , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/fisiología , Animales , Células Cultivadas , Quimiocina CCL2/antagonistas & inhibidores , Interleucina-1beta/biosíntesis , Ratones , Miocitos del Músculo Liso/inmunología , FN-kappa B/fisiología , Receptores CCR2/antagonistas & inhibidores , Proteína Smad4/fisiología , Tamoxifeno/farmacología
17.
Immunity ; 46(4): 635-648, 2017 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-28410990

RESUMEN

Mice carrying a hypomorphic point mutation in the Ptpn6 gene (Ptpn6spin mice) develop an inflammatory skin disease that resembles neutrophilic dermatosis in humans. Here, we demonstrated that interleukin-1α (IL-1α) signaling through IL-1R and MyD88 in both stromal and immune cells drive inflammation in Ptpn6spin mice. We further identified SYK as a critical kinase that phosphorylates MyD88, promoted MyD88-dependent signaling and mediates dermatosis in Ptpn6spin mice. Our studies further demonstrated that SHP1 encoded by Ptpn6 binds and suppresses SYK activation to inhibit MyD88 phosphorylation. Downstream of SHP1 and SYK-dependent counterregulation of MyD88 tyrosine phosphorylation, we have demonstrated that the scaffolding function of receptor interacting protein kinase 1 (RIPK1) and tumor growth factor-ß activated kinase 1 (TAK1)-mediating signaling were required to spur inflammatory disease. Overall, these studies identify SHP1 and SYK crosstalk as a critical regulator of MyD88 post-translational modifications and IL-1-driven inflammation.


Asunto(s)
Inflamación/inmunología , Interleucina-1alfa/inmunología , Factor 88 de Diferenciación Mieloide/inmunología , Enfermedades de la Piel/inmunología , Quinasa Syk/inmunología , Animales , Citometría de Flujo , Células HEK293 , Humanos , Immunoblotting , Inflamación/genética , Inflamación/metabolismo , Interleucina-1alfa/genética , Interleucina-1alfa/metabolismo , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/inmunología , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones Noqueados , Modelos Inmunológicos , Mutación , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/inmunología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Receptores de Interleucina-1/inmunología , Receptores de Interleucina-1/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Enfermedades de la Piel/genética , Enfermedades de la Piel/metabolismo , Quinasa Syk/genética , Quinasa Syk/metabolismo
18.
Clin Microbiol Rev ; 37(2): e0007323, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38421182

RESUMEN

SUMMARYViral infections during pregnancy are associated with significant adverse perinatal and fetal outcomes. Pregnancy is a unique immunologic and physiologic state, which can influence control of virus replication, severity of disease, and vertical transmission. The placenta is the organ of the maternal-fetal interface and provides defense against microbial infection while supporting the semi-allogeneic fetus via tolerogenic immune responses. Some viruses, such as cytomegalovirus, Zika virus, and rubella virus, can breach these defenses, directly infecting the fetus and having long-lasting consequences. Even without direct placental infection, other viruses, including respiratory viruses like influenza viruses and severe acute respiratory syndrome coronavirus 2, still cause placental damage and inflammation. Concentrations of progesterone and estrogens rise during pregnancy and contribute to immunological adaptations, placentation, and placental development and play a pivotal role in creating a tolerogenic environment at the maternal-fetal interface. Animal models, including mice, nonhuman primates, rabbits, and guinea pigs, are instrumental for mechanistic insights into the pathogenesis of viral infections during pregnancy and identification of targetable treatments to improve health outcomes of pregnant individuals and offspring.


Asunto(s)
Complicaciones Infecciosas del Embarazo , Virosis , Embarazo , Femenino , Humanos , Complicaciones Infecciosas del Embarazo/inmunología , Complicaciones Infecciosas del Embarazo/virología , Animales , Virosis/inmunología , Virosis/transmisión , Placenta/virología , Placenta/inmunología , Transmisión Vertical de Enfermedad Infecciosa , Modelos Animales de Enfermedad
19.
Dev Biol ; 510: 8-16, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38403101

RESUMEN

Physiological root resorption is a common occurrence during the development of deciduous teeth in children. Previous research has shown that the regulation of the inflammatory microenvironment through autophagy in DDPSCs is a significant factor in this process. However, it remains unclear why there are variations in the autophagic status of DDPSCs at different stages of physiological root resorption. To address this gap in knowledge, this study examines the relationship between the circadian clock of DDPSCs, the autophagic status, and the periodicity of masticatory behavior. Samples were collected from deciduous teeth at various stages of physiological root resorption, and DDPSCs were isolated and cultured for analysis. The results indicate that the circadian rhythm of important autophagy genes, such as Beclin-1 and LC3, and the clock gene REV-ERBα in DDPSCs, disappears under mechanical stress. Additionally, the study found that REV-ERBα can regulate Beclin-1 and LC3. Evidence suggests that mechanical stress is a trigger for the regulation of autophagy via REV-ERBα. Overall, this study highlights the importance of mechanical stress in regulating autophagy of DDPSCs via REV-ERBα, which affects the formation of the inflammatory microenvironment and plays a critical role in physiological root resorption in deciduous teeth.


Asunto(s)
Relojes Circadianos , Resorción Radicular , Niño , Humanos , Resorción Radicular/genética , Beclina-1/genética , Ritmo Circadiano/genética , Células Madre , Diente Primario
20.
Immunity ; 45(6): 1258-1269, 2016 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-27939674

RESUMEN

Programmed death and shedding of epithelial cells is a powerful defense mechanism to reduce bacterial burden during infection but this activity cannot be indiscriminate because of the critical barrier function of the epithelium. We report that during cystitis, shedding of infected bladder epithelial cells (BECs) was preceded by the recruitment of mast cells (MCs) directly underneath the superficial epithelium where they docked and extruded their granules. MCs were responding to interleukin-1ß (IL-1ß) secreted by BECs after inflammasome and caspase-1 signaling. Upon uptake of granule-associated chymase (mouse MC protease 4 [mMCPT4]), BECs underwent caspase-1-associated cytolysis and exfoliation. Thus, infected epithelial cells require a specific cue for cytolysis from recruited sentinel inflammatory cells before shedding.


Asunto(s)
Quimasas/inmunología , Citotoxinas/inmunología , Células Epiteliales/microbiología , Mastocitos/inmunología , Infecciones Urinarias/inmunología , Animales , Degranulación de la Célula/inmunología , Línea Celular , Gránulos Citoplasmáticos/química , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA