Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Nat Immunol ; 13(12): 1162-70, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23086447

RESUMEN

The NF-κB protein RelB controls dendritic cell (DC) maturation and may be targeted therapeutically to manipulate T cell responses in disease. Here we report that RelB promoted DC activation not as the expected RelB-p52 effector of the noncanonical NF-κB pathway, but as a RelB-p50 dimer regulated by canonical IκBs, IκBα and IκBɛ. IκB control of RelB minimized spontaneous maturation but enabled rapid pathogen-responsive maturation. Computational modeling of the NF-κB signaling module identified control points of this unexpected cell type-specific regulation. Fibroblasts that we engineered accordingly showed DC-like RelB control. Canonical pathway control of RelB regulated pathogen-responsive gene expression programs. This work illustrates the potential utility of systems analyses in guiding the development of combination therapeutics for modulating DC-dependent T cell responses.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Activación de Linfocitos , FN-kappa B/metabolismo , Factor de Transcripción ReIB/metabolismo , Animales , Diferenciación Celular/genética , Línea Celular , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Quinasa I-kappa B/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/genética , Multimerización de Proteína , Transducción de Señal , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 9/metabolismo , Factor de Transcripción ReIB/genética
2.
Proc Natl Acad Sci U S A ; 111(38): 13942-7, 2014 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-25201978

RESUMEN

Mammalian hosts are colonized with commensal microbes in various mucosal and epithelial tissues, including the intestinal tract. In mice, the presence of segmented filamentous bacteria (SFB) promotes Th17 differentiation and the development of autoimmune disease. Here, we demonstrate that the IL-23 pathway dynamically regulates the abundance of SFB as well as mucosal barrier function in the adult animal. Genetic or pharmacological inactivation of the pathway selectively perturbs the abundance of a small group of commensals, including SFB, and results in an impaired mucosal barrier. Defective barrier function leads to systemic dissemination of microbial products, provoking induction of the IL-23 pathway with dual consequences: IL-23 drives IL-22 production to reinforce mucosal barrier function and elicit antimicrobial activities, and it also drives the differentiation of Th17 cells in an attempt to combat escaped microbes in the lamina propria and in distal tissues. Thus, barrier defects generate a systemic environment that facilitates Th17 development.


Asunto(s)
Interleucinas/inmunología , Mucosa Intestinal/inmunología , Microbiota/inmunología , Receptores de Interleucina/inmunología , Células Th17/inmunología , Animales , Diferenciación Celular/inmunología , Interleucinas/genética , Mucosa Intestinal/microbiología , Ratones , Ratones Noqueados , Receptores de Interleucina/genética , Interleucina-22
3.
Proc Natl Acad Sci U S A ; 106(24): 9619-24, 2009 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-19487661

RESUMEN

Mammalian signaling networks contain an abundance of negative feedback regulators that may have overlapping ("fail-safe") or specific functions. Within the NF-kappaB signaling module, IkappaB alpha is known as a negative feedback regulator, but the newly characterized inhibitor IkappaB delta is also inducibly expressed in response to inflammatory stimuli. To examine IkappaB delta's roles in inflammatory signaling, we mathematically modeled the 4-IkappaB-containing NF-kappaB signaling module and developed a computational phenotyping methodology of general applicability. We found that IkappaB delta, like IkappaB alpha, can provide negative feedback, but each functions stimulus-specifically. Whereas IkappaB delta attenuates persistent, pathogen-triggered signals mediated by TLRs, the more prominent IkappaB alpha does not. Instead, IkappaB alpha, which functions more rapidly, is primarily involved in determining the temporal profile of NF-kappaB signaling in response to cytokines that serve intercellular communication. Indeed, when removing the inducing cytokine stimulus by compound deficiency of the tnf gene, we found that the lethality of ikappab alpha(-/-) mouse was rescued. Finally, we found that IkappaB delta provides signaling memory owing to its long half-life; it integrates the inflammatory history of the cell to dampen NF-kappaB responsiveness during sequential stimulation events.


Asunto(s)
FN-kappa B/metabolismo , Receptores de Citocinas/metabolismo , Transducción de Señal , Factor de Transcripción ReIA/metabolismo , Células 3T3 , Animales , Cinética , Ratones , Ratones Endogámicos C57BL
4.
Mol Cancer Ther ; 18(4): 780-787, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30824607

RESUMEN

To provide a better understanding of the pharmacokinetics-pharmacodynamics relationships of antibody-based drugs, we analyzed several chimeric and humanized monoclonal antibodies or antibody-drug conjugates (ADC) for PK and efficacy among four strains of mice. Notably, antibodies and ADCs displayed a dose-dependent drug disposition profile in the plasma of NSG mice. The increased clearance rate in NSG mice resulted in the reduction of antitumor activity of ADCs. Furthermore, we identified that the abnormal clearance was mediated by Fc-FcγR interaction by comparing antibodies that lack FcγR binding capacity. We also found a high percentage of FcγR-expressing macrophages in the bone marrow, spleen, and liver of NSG mice, which may be responsible for the abnormal distribution of antibodies. Overall, these findings suggest that preclinical evaluation of efficacy and pharmacokinetics of antibodies and ADCs need to consider mouse strain-induced variations.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Inmunoconjugados/uso terapéutico , Linfoma Anaplásico de Células Grandes/tratamiento farmacológico , Receptores de IgG/metabolismo , Animales , Anticuerpos Monoclonales/metabolismo , Antineoplásicos/metabolismo , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Femenino , Semivida , Humanos , Inmunoconjugados/metabolismo , Antígeno Ki-1/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Células Mieloides/inmunología , Células Mieloides/metabolismo , Glutamato de Sodio/farmacología , Resultado del Tratamiento , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
5.
J Invest Dermatol ; 135(8): 2012-2020, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25815426

RESUMEN

The inflammasome is a complex of proteins that has a critical role in mounting an inflammatory response in reply to a harmful stimulus that compromises the homeostatic state of the tissue. The NLRP3 inflammasome, which is found in a wound-like environment, is comprised of three components: the NLRP3, the adaptor protein ASC and caspase-1. Interestingly, although ASC levels do not fluctuate, caspase-1 levels are elevated in both physiological and pathological conditions. Despite the observation that merely raising caspase-1 levels is sufficient to induce inflammation, the crucial question regarding the mechanism governing its expression is unexplored. We found that, in an inflammatory microenvironment, caspase-1 is regulated by NF-κB. Consistent with this association, the inhibition of caspase-1 activity parallels the effects on wound healing caused by the abrogation of NF-κB activation. Surprisingly, not only does inhibition of the NF-κB/caspase-1 axis disrupt the inflammatory phase of the wound-healing program, but it also impairs the stimulation of cutaneous epithelial stem cells of the proliferative phase. These data provide a mechanistic basis for the complex interplay between different phases of the wound-healing response in which the downstream signaling activity of immune cells can kindle the amplification of local stem cells to advance tissue repair.


Asunto(s)
Caspasa 1/fisiología , Microambiente Celular/fisiología , Dermatitis/patología , Dermatitis/fisiopatología , Queratinocitos/patología , Animales , Caspasa 8/genética , Caspasa 8/fisiología , Proliferación Celular/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Homeostasis/fisiología , Técnicas In Vitro , Ratones , Ratones Noqueados , FN-kappa B/genética , FN-kappa B/fisiología , Transducción de Señal/fisiología , Cicatrización de Heridas/fisiología
6.
Cancer Cell ; 24(2): 151-66, 2013 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-23850221

RESUMEN

IκB proteins are the primary inhibitors of NF-κB. Here, we demonstrate that sumoylated and phosphorylated IκBα accumulates in the nucleus of keratinocytes and interacts with histones H2A and H4 at the regulatory region of HOX and IRX genes. Chromatin-bound IκBα modulates Polycomb recruitment and imparts their competence to be activated by TNFα. Mutations in the Drosophila IκBα gene cactus enhance the homeotic phenotype of Polycomb mutants, which is not counteracted by mutations in dorsal/NF-κB. Oncogenic transformation of keratinocytes results in cytoplasmic IκBα translocation associated with a massive activation of Hox. Accumulation of cytoplasmic IκBα was found in squamous cell carcinoma (SCC) associated with IKK activation and HOX upregulation.


Asunto(s)
Cromatina/metabolismo , Proteínas I-kappa B/metabolismo , Neoplasias Cutáneas/metabolismo , Animales , Diferenciación Celular/genética , Núcleo Celular/genética , Núcleo Celular/metabolismo , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Cromatina/genética , Células HEK293 , Histonas/genética , Histonas/metabolismo , Humanos , Proteínas I-kappa B/genética , Queratinocitos/citología , Queratinocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Inhibidor NF-kappaB alfa , Transducción de Señal , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología
7.
Cell Res ; 21(1): 86-102, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21102550

RESUMEN

Two distinct nuclear factor κB (NFκB) signaling pathways have been described; the canonical pathway that mediates inflammatory responses, and the non-canonical pathway that is involved in immune cell differentiation and maturation and secondary lymphoid organogenesis. The former is dependent on the IκB kinase adaptor molecule NEMO, the latter is independent of it. Here, we review the molecular mechanisms of regulation in each signaling axis and attempt to relate the apparent regulatory logic to the physiological function. Further, we review the recent evidence for extensive cross-regulation between these two signaling axes and summarize them in a wiring diagram. These observations suggest that NEMO-dependent and -independent signaling should be viewed within the context of a single NFκB signaling system, which mediates signaling from both inflammatory and organogenic stimuli in an integrated manner. As in other regulatory biological systems, a systems approach including mathematical models that include quantitative and kinetic information will be necessary to characterize the network properties that mediate physiological function, and that may break down to cause or contribute to pathology.


Asunto(s)
FN-kappa B/metabolismo , Humanos , Quinasa I-kappa B/metabolismo , Inflamación/metabolismo , Proteínas Inhibidoras de la Apoptosis/metabolismo , FN-kappa B/fisiología , Transducción de Señal , Factor 2 Asociado a Receptor de TNF/metabolismo , Factor 3 Asociado a Receptor de TNF/metabolismo
8.
Mol Cell Biol ; 28(10): 3139-50, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18299388

RESUMEN

The NF-kappaB signaling pathway regulates the activity of multiple dimeric transcription factors that are generated from five distinct monomers. The availabilities of specific dimers are regulated during cell differentiation and organ development and determine the cell's responsiveness to inflammatory or developmental signals. An altered dimer distribution is a hallmark of many chronic diseases. Here, we reveal that the cellular processes that generate different NF-kappaB dimers are highly connected through multiple cross-regulatory mechanisms. First, we find that steady-state expression of RelB is regulated by the canonical pathway and constitutive RelA activity. Indeed, synthesis control of RelB is the major determinant of noncanonical NF-kappaB dimer activation. Second, processing, not synthesis, of p100 and p105 is mechanistically linked via competitive dimerization with a limited pool of RelA and RelB. This homeostatic cross-regulatory mechanism determines the availability of the p50- and p52-containing dimers and also of the noncanonical IkappaB p100. Our results inform a wiring diagram to delineate NF-kappaB dimer formation that emphasizes that inflammatory and developmental signaling cannot be considered separately but are highly interconnected.


Asunto(s)
FN-kappa B/metabolismo , Factores de Transcripción/metabolismo , Animales , Células Cultivadas , Dimerización , Quinasa I-kappa B/deficiencia , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Ganglios Linfáticos/crecimiento & desarrollo , Ganglios Linfáticos/metabolismo , Ratones , Ratones Noqueados , FN-kappa B/genética , Subunidad p50 de NF-kappa B/deficiencia , Subunidad p50 de NF-kappa B/genética , Subunidad p50 de NF-kappa B/metabolismo , Subunidad p52 de NF-kappa B/deficiencia , Subunidad p52 de NF-kappa B/genética , Subunidad p52 de NF-kappa B/metabolismo , Estructura Cuaternaria de Proteína , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Factor de Transcripción ReIA/química , Factor de Transcripción ReIA/deficiencia , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Factor de Transcripción ReIB/química , Factor de Transcripción ReIB/deficiencia , Factor de Transcripción ReIB/genética , Factor de Transcripción ReIB/metabolismo , Factores de Transcripción/química , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA