Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 13: 913275, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36110848

RESUMEN

Activation of CD40-signaling contributes to the initiation, progression and drug resistance of B cell lymphomas. We contributed to this knowledge by showing that constitutive CD40-signaling in B cells induces B cell hyperplasia and finally B cell lymphoma development in transgenic mice. CD40 activates, among others, the non-canonical NF-ĸB signaling, which is constitutively activated in several human B cell lymphomas and is therefore presumed to contribute to lymphopathogenesis. This prompted us to study the regulatory role of the non-canonical NF-ĸB transcription factor RelB in lymphomagenesis. To this end, we crossed mice expressing a constitutively active CD40 receptor in B cells with conditional RelB-KO mice. Ablation of RelB attenuated pre-malignant B cell expansion, and resulted in an impaired survival and activation of long-term CD40-stimulated B cells. Furthermore, we found that hyperactivation of non-canonical NF-кB signaling enhances the retention of B cells in the follicles of secondary lymphoid organs. RNA-Seq-analysis revealed that several genes involved in B-cell migration, survival, proliferation and cytokine signaling govern the transcriptional differences modulated by the ablation of RelB in long-term CD40-stimulated B cells. Inactivation of RelB did not abrogate lymphoma development. However, lymphomas occurred with a lower incidence and had a longer latency period. In summary, our data suggest that RelB, although it is not strictly required for malignant transformation, accelerates the lymphomagenesis of long-term CD40-stimulated B cells by regulating genes involved in migration, survival and cytokine signaling.


Asunto(s)
Linfoma de Células B , Linfoma , Factor de Transcripción ReIB , Animales , Linfocitos B , Antígenos CD40/genética , Citocinas , Humanos , Linfoma de Células B/genética , Ratones , Ratones Transgénicos , FN-kappa B , Factor de Transcripción ReIB/genética
2.
J Immunol ; 203(10): 2602-2613, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31578269

RESUMEN

Foxp3+ regulatory T cells are well-known immune suppressor cells in various settings. In this study, we provide evidence that knockout of the relB gene in dendritic cells (DCs) of C57BL/6 mice results in a spontaneous and systemic accumulation of Foxp3+ T regulatory T cells (Tregs) partially at the expense of microbiota-reactive Tregs. Deletion of nfkb2 does not fully recapitulate this phenotype, indicating that alternative NF-κB activation via the RelB/p52 complex is not solely responsible for Treg accumulation. Deletion of RelB in DCs further results in an impaired oral tolerance induction and a marked type 2 immune bias among accumulated Foxp3+ Tregs reminiscent of a tissue Treg signature. Tissue Tregs were fully functional, expanded independently of IL-33, and led to an almost complete Treg-dependent protection from experimental autoimmune encephalomyelitis. Thus, we provide clear evidence that RelB-dependent pathways regulate the capacity of DCs to quantitatively and qualitatively impact on Treg biology and constitute an attractive target for treatment of autoimmune diseases but may come at risk for reduced immune tolerance in the intestinal tract.


Asunto(s)
Autoinmunidad/genética , Células Dendríticas/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Linfocitos T Reguladores/inmunología , Factor de Transcripción ReIB/metabolismo , Animales , Células Cultivadas , Factores de Transcripción Forkhead/metabolismo , Técnicas de Inactivación de Genes , Homeostasis/inmunología , Tolerancia Inmunológica/inmunología , Inflamación/inmunología , Interleucina-33/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Subunidad p52 de NF-kappa B/metabolismo , Factor de Transcripción ReIB/deficiencia , Factor de Transcripción ReIB/genética
3.
Immunobiology ; 224(5): 687-696, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31200979

RESUMEN

Regulatory T cells (Tregs) maintain immune homeostasis and play an important role in tissue regeneration after injury. Mutations affecting development or homeostasis of Tregs lead to immune pathologies in humans and are often fatal in mouse models. Although the pathways required for Treg development are being increasingly characterized, factors crucial for Treg homeostasis are not completely understood. Previously we have found a role for alternative NF-κB pathway in restricting T cell activation and Th17 differentiation. Here, by using the mouse model of uncontrolled alternative NF-κB signaling we identify a crucial intrinsic role of RelB signaling in regulating homeostasis and competitive fitness of Tregs. The failure of p100-/- Tregs to maintain the population of effector Tregs and efficiently suppress immune reactions results in lethal multiorgan Th1-mediated inflammation in Rag1-/- recipients. This inflammation is combined with severe lymphopenia and could be rescued by adoptive transfer of wild type Tregs. Thus in addition to its role in Th17 differentiation, RelB acts as a potent inhibitor of Treg effector functions. Our results point to RelB as a potential therapeutic target for Treg manipulation.


Asunto(s)
Homeostasis , FN-kappa B/metabolismo , Transducción de Señal , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Animales , Autoinmunidad , Biomarcadores , Citocinas/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Inmunohistoquímica , Inmunomodulación/genética , Inmunofenotipificación , Activación de Linfocitos , Ratones , Ratones Noqueados , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Factor de Transcripción ReIB/metabolismo , Proteína Activadora de GTPasa p120/genética , Proteína Activadora de GTPasa p120/metabolismo
4.
Bone ; 121: 29-41, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30611922

RESUMEN

Endochondral ossification is important for skeletal development. Recent findings indicate that the p65 (RelA) subunit, a main subunit of the classical nuclear factor-κB (NF-κB) pathway, plays essential roles in chondrocyte differentiation. Although several groups have reported that the alternative NF-κB pathway also regulates bone homeostasis, the role of the alternative NF-κB pathway in chondrocyte development is still unclear. Here, we analyzed the in vivo function of the alternative pathway on endochondral ossification using p100-deficient (p100-/-) mice, which carry a homozygous deletion of the COOH-terminal ankyrin repeats of p100 but still express functional p52 protein. The alternative pathway was activated during the periarticular stage in wild-type mice. p100-/- mice exhibited dwarfism, and histological analysis of the growth plate revealed abnormal arrangement of chondrocyte columns and a narrowed hypertrophic zone. Consistent with these observations, the expression of hypertrophic chondrocyte markers, type X collagen (ColX) or matrix metalloproteinase 13, but not early chondrogenic markers, such as Col II or aggrecan, was suppressed in p100-/- mice. An in vivo BrdU tracing assay clearly demonstrated less proliferative activity in chondrocytes in p100-/- mice. These defects were partly rescued when the RelB gene was deleted in p100-/- mice. Taken together, the alternative NF-κB pathway may regulate chondrocyte proliferation and differentiation to maintain endochondral ossification.


Asunto(s)
FN-kappa B/metabolismo , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Proliferación Celular/genética , Proliferación Celular/fisiología , Condrocitos/metabolismo , Condrogénesis/genética , Condrogénesis/fisiología , Inmunohistoquímica , Hibridación in Situ , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteogénesis/genética , Osteogénesis/fisiología , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/genética , Transducción de Señal/fisiología , Esqueleto/metabolismo
5.
Eur J Immunol ; 48(6): 923-936, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29485182

RESUMEN

The NF-κB transcription factor subunit RelB is important for the full activation of conventional dendritic cells (cDCs) during T-cell-dependent immune responses. Although the number of splenic DCs is greatly reduced in RelBnull mice, the cause and consequences of this deficiency are currently unknown. To circumvent the impact of the pleiotropic defects in RelBnull mice we used a reporter model for RelB expression (RelBKatushka mice) and conditionally deleted RelB in DCs (RelBCD11c-Cre mice). Thereby, we can show here that RelB is essential for the differentiation of a CD117+ CD172a+ cDC subpopulation that highly expresses RelB. Surprisingly, these DCs depend on p50 for their development and are negatively regulated by a constitutive p52 activation in absence of p100. The absence of p52/p100 had no influence on the homeostasis of CD117+ CD172a+ cDCs. RelB-dependent CD117+ CD172a+ DCs strongly induce the production of the type 2 cytokines IL-4 and IL-13, as well as GM-CSF from naïve Th cells. Consequently, mice lacking RelB in cDCs show an attenuated bronchial hyperresponsiveness with reduced eosinophil infiltration. Taken together, we have identified a new splenic RelB-dependent CD117+ CD172a+ cDC population that preferentially induces Th2 responses.


Asunto(s)
Hiperreactividad Bronquial/inmunología , Células Dendríticas/fisiología , Eosinófilos/inmunología , Subunidad p50 de NF-kappa B/metabolismo , Células Th2/inmunología , Factor de Transcripción ReIB/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Citocinas/metabolismo , Regulación de la Expresión Génica , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas c-kit/metabolismo , Receptores Inmunológicos/metabolismo , Factor de Transcripción ReIB/genética
6.
Immunobiology ; 223(2): 191-199, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29050819

RESUMEN

The role of the alternative NF-κB pathway is mainly attributed to the lymphoid organ formation and blood cancer. However, its involvement in lymphocyte differentiation is not clearly defined. Recently, we have shown that uncontrolled activation of alternative NF-κB in mice lacking the NF-κB inhibitory protein p100 (p100-/- mice) hinders plasmablast proliferation and diminishes T cell independent responses. Here we show that hyperactivation of this pathway leads to a cell-intrinsic T cell defects. p100-deficient T helper cells displayed both an activation and a proliferation defect in vitro. In addition, memory T cell formation was impaired in vivo. Moreover, p100-/- T cells failed to polarize into T helper 17 cells. This phenotype was dependent on increased RelB activation and suboptimal RORγt expression. Thus, our results demonstrate that RelB acts as a negative regulator of T cell activation and Th17 development. Targeting this pathway therefore could be beneficial in Th17-mediated pathologies.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Linfocitos B/fisiología , Inflamación/inmunología , Células Plasmáticas/fisiología , Subgrupos de Linfocitos T/inmunología , Células Th17/inmunología , Factor de Transcripción ReIB/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Regulación hacia Abajo , Memoria Inmunológica , Activación de Linfocitos , Ratones , Ratones Noqueados , Subunidad p52 de NF-kappa B/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Especificidad de Órganos , Factor de Transcripción ReIB/genética
7.
Geroscience ; 39(5-6): 557-570, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28891034

RESUMEN

The mechanistic target of rapamycin (mTOR), a protein kinase, is a central regulator of mammalian metabolism and physiology. Protein mTOR complex 1 (mTORC1) functions as a major sensor for the nutrient, energy, and redox state of a cell and is activated by ras homolog enriched in brain (RHEB1), a GTP-binding protein. Increased activation of mTORC1 pathway has been associated with developmental abnormalities, certain form of epilepsy (tuberous sclerosis), and cancer. Clinically, those mTOR-related disorders are treated with the mTOR inhibitor rapamycin and its rapalogs. Because the effects of chronic interference with mTOR signaling in the aged brain are yet unknown, we used a genetic strategy to interfere with mTORC1 signaling selectively by introducing mutations of Rheb1 into the mouse. We created conventional knockout (Rheb1 +/- ) and gene trap (Rheb1 Δ/+ ) mutant mouse lines. Rheb1-insufficient mice with different combinations of mutant alleles were monitored over a time span of 2 years. The mice did not show any behavioral/neurological changes during the first 18 months of age. However, after aging (> 18 months of age), both the Rheb1 +/- and Rheb1 Δ /- hybrid males developed rare stress-induced seizures, whereas Rheb1 +/- and Rheb1 Δ /- females and Rheb1 Δ/+ and Rheb1 Δ/Δ mice of both genders did not show any abnormality. Our findings suggest that chronic intervention with mTORC1 signaling in the aged brain might be associated with major adverse events.


Asunto(s)
Envejecimiento/genética , Proteína Homóloga de Ras Enriquecida en el Cerebro/deficiencia , Convulsiones/etiología , Estrés Psicológico/genética , Animales , Conducta Animal , Western Blotting/métodos , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Masculino , Ratones , Terapia Molecular Dirigida/métodos , Fenotipo , ARN Mensajero/análisis , ARN Mensajero/genética , Distribución Aleatoria , Proteína Homóloga de Ras Enriquecida en el Cerebro/genética , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Valores de Referencia , Convulsiones/genética , Transducción de Señal , Estrés Psicológico/complicaciones
8.
Front Immunol ; 8: 726, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28690613

RESUMEN

Thymus-derived natural Foxp3+ CD4+ regulatory T cells (nTregs) play a key role in maintaining immune tolerance and preventing autoimmune disease. Several studies indicate that dendritic cells (DCs) are critically involved in the maintenance and proliferation of nTregs. However, the mechanisms how DCs manage to keep the peripheral pool at constant levels remain poorly understood. Here, we describe that the NF-κB/Rel family transcription factor RelB controls the frequencies of steady-state migratory DCs (ssmDCs) in peripheral lymph nodes and their numbers control peripheral nTreg homeostasis. DC-specific RelB depletion was investigated in CD11c-Cre × RelBfl/fl mice (RelBDCko), which showed normal frequencies of resident DCs in lymph nodes and spleen while the subsets of CD103- Langerin- dermal DCs (dDCs) and Langerhans cells but not CD103+ Langerin+ dDC of the ssmDCs in skin-draining lymph nodes were increased. Enhanced frequencies and proliferation rates were also observed for nTregs and a small population of CD4+ CD44high CD25low memory-like T cells (Tml). Interestingly, only the Tml but not DCs showed an increase in IL-2-producing capacity in lymph nodes of RelBDCko mice. Blocking of IL-2 in vivo reduced the frequency of nTregs but increased the Tml frequencies, followed by a recovery of nTregs. Taken together, by employing RelBDCko mice with increased frequencies of ssmDCs our data indicate a critical role for specific ssmDC subsets for the peripheral nTreg and IL-2+ Tml frequencies during homeostasis.

9.
J Autoimmun ; 81: 56-67, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28385374

RESUMEN

Medullary thymic epithelial cells (mTECs) contribute to self-tolerance by expressing and presenting peripheral tissue antigens for negative selection of autoreactive T cells and differentiation of natural regulatory T cells. The molecular control of mTEC development remains incompletely understood. We here demonstrate by TEC-specific gene manipulation in mice that the NF-κB transcription factor subunit RelB, which is activated by the alternative NF-κB pathway, regulates development of mature mTECs in a dose-dependent manner. Mice with conditional deletion of Relb lacked mature mTECs and developed spontaneous autoimmunity. In addition, the NF-κB subunits RelA and c-Rel, which are both activated by classical NF-κB signaling, were jointly required for mTEC differentiation by directly regulating the transcription of Relb. Our data reveal a crosstalk mechanism between classical and alternative NF-κB pathways that tightly controls the development of mature mTECs to ensure self-tolerance.


Asunto(s)
Tolerancia Central/inmunología , Células Epiteliales/metabolismo , FN-kappa B/metabolismo , Transducción de Señal , Timo/inmunología , Timo/metabolismo , Animales , Autoinmunidad/genética , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Línea Celular , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Epiteliales/citología , Expresión Génica , Hígado/inmunología , Hígado/metabolismo , Hígado/patología , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Noqueados , Ratones Transgénicos , FN-kappa B/genética , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo
10.
Immunity ; 44(3): 553-567, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26982364

RESUMEN

Intestinal epithelial cells (IECs) regulate gut immune homeostasis, and impaired epithelial responses are implicated in the pathogenesis of inflammatory bowel diseases (IBD). IEC-specific ablation of nuclear factor κB (NF-κB) essential modulator (NEMO) caused Paneth cell apoptosis and impaired antimicrobial factor expression in the ileum, as well as colonocyte apoptosis and microbiota-driven chronic inflammation in the colon. Combined RelA, c-Rel, and RelB deficiency in IECs caused Paneth cell apoptosis but not colitis, suggesting that NEMO prevents colon inflammation by NF-κB-independent functions. Inhibition of receptor-interacting protein kinase 1 (RIPK1) kinase activity or combined deficiency of Fas-associated via death domain protein (FADD) and RIPK3 prevented epithelial cell death, Paneth cell loss, and colitis development in mice with epithelial NEMO deficiency. Therefore, NEMO prevents intestinal inflammation by inhibiting RIPK1 kinase activity-mediated IEC death, suggesting that RIPK1 inhibitors could be effective in the treatment of colitis in patients with NEMO mutations and possibly in IBD.


Asunto(s)
Enfermedades Inflamatorias del Intestino/inmunología , Mucosa Intestinal/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Células de Paneth/fisiología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Animales , Apoptosis/genética , Células Cultivadas , Proteína de Dominio de Muerte Asociada a Fas/genética , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-rel/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIB/genética
11.
Cancer Cell ; 28(5): 582-598, 2015 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-26555174

RESUMEN

IκB kinase/nuclear [corrected] factor κB (IKK/NF-κB) signaling exhibits important yet opposing functions in hepatocarcinogenesis. Mice lacking NEMO in liver parenchymal cells (LPC) spontaneously develop steatohepatitis and hepatocellular carcinoma (HCC) suggesting that NF-κB prevents liver disease and cancer. Here, we show that complete NF-κB inhibition by combined LPC-specific ablation of RelA, c-Rel, and RelB did not phenocopy NEMO deficiency, but constitutively active IKK2-mediated NF-κB activation prevented hepatocellular damage and HCC in NEMO(LPC-KO) mice. Knock-in expression of kinase inactive receptor-interacting protein kinase 1 (RIPK1) prevented hepatocyte apoptosis and HCC, while RIPK1 ablation induced TNFR1-associated death domain protein (TRADD)-dependent hepatocyte apoptosis and liver tumors in NEMO(LPC-KO) mice, revealing distinct kinase-dependent and scaffolding functions of RIPK1. Collectively, these results show that NEMO prevents hepatocarcinogenesis by inhibiting RIPK1 kinase activity-driven hepatocyte apoptosis through NF-κB-dependent and -independent functions.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Hígado Graso/metabolismo , Hepatocitos/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias Hepáticas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Animales , Apoptosis/genética , Carcinoma Hepatocelular/genética , Células Cultivadas , Hígado Graso/genética , Expresión Génica , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Immunoblotting , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intracelular/genética , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas/genética , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/genética , FN-kappa B/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética
12.
Methods Mol Biol ; 1339: 297-308, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26445797

RESUMEN

Atherosclerosis is a transmural chronic inflammatory disease of medium and large arteries. Though it is well recognized that immune responses contribute to atherosclerosis, it remains unclear whether these responses are carried out in secondary lymphoid organs such as the spleen and lymph nodes and/or within the arterial wall. Arteries are composed of three major layers, i.e., the laminae intima, media, and adventitia. However, each of these layers may play different roles in arterial wall biology and atherogenesis. We identified well-structured artery tertiary lymphoid organs (ATLOs) in the abdominal aorta adventitia but not in the intima of aged apolipoprotein E-null (ApoE(-/-)) mice. These observations suggested that disease-associated immune responses are highly territorialized within the arterial wall and that the adventitia may play distinct and hitherto unrecognized roles. Here, we set out to apply laser capture microdissection (LCM) to dissect plaque, media, adventitia, and adjacent aorta-draining lymph nodes (LN) in aged ApoE(-/-) mice in attempts to establish the territoriality of atherosclerosis immune responses. Using whole-genome mRNA expression microarrays of arterial wall tissues, we constructed robust transcript atlases of wild-type and ApoE(-/-) mouse aortas. Data were deposited in the National Center for Biotechnology Information's gene expression omnibus (GEO) and are accessible to the public through the Internet. These transcript atlases are anticipated to prove valuable to address a wide scope of issues ranging from atherosclerosis immunity and inflammation to the role of single genes in regulating arterial wall remodeling. This chapter presents protocols for LCM of mouse aorta and microarray expression analysis from LCM-isolated aorta laminae.


Asunto(s)
Aorta/metabolismo , Enfermedades de la Aorta/genética , Apolipoproteínas E/deficiencia , Aterosclerosis/genética , Perfilación de la Expresión Génica/métodos , Captura por Microdisección con Láser , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , Acceso a la Información , Animales , Enfermedades de la Aorta/inmunología , Enfermedades de la Aorta/metabolismo , Apolipoproteínas E/genética , Aterosclerosis/inmunología , Aterosclerosis/metabolismo , Biología Computacional , Bases de Datos Genéticas , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Internet , Ratones Noqueados , Fenotipo , Transcriptoma
13.
Nat Commun ; 6: 7796, 2015 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-26183376

RESUMEN

Acute lung injury (ALI) is a severe inflammatory disease for which no specific treatment exists. As glucocorticoids have potent immunosuppressive effects, their application in ALI is currently being tested in clinical trials. However, the benefits of this type of regimen remain unclear. Here we identify a mechanism of glucocorticoid action that challenges the long-standing dogma of cytokine repression by the glucocorticoid receptor. Contrarily, synergistic gene induction of sphingosine kinase 1 (SphK1) by glucocorticoids and pro-inflammatory stimuli via the glucocorticoid receptor in macrophages increases circulating sphingosine 1-phosphate levels, which proves essential for the inhibition of inflammation. Chemical or genetic inhibition of SphK1 abrogates the therapeutic effects of glucocorticoids. Inflammatory p38 MAPK- and mitogen- and stress-activated protein kinase 1 (MSK1)-dependent pathways cooperate with glucocorticoids to upregulate SphK1 expression. Our findings support a critical role for SphK1 induction in the suppression of lung inflammation by glucocorticoids, and therefore provide rationales for effective anti-inflammatory therapies.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Glucocorticoides/farmacología , Macrófagos/efectos de los fármacos , Fosfotransferasas (Aceptor de Grupo Alcohol)/efectos de los fármacos , Receptores de Glucocorticoides/agonistas , Animales , Inmunoprecipitación de Cromatina , Citocinas/efectos de los fármacos , Citocinas/inmunología , Citometría de Flujo , Regulación de la Expresión Génica/efectos de los fármacos , Inflamación , Lisofosfolípidos/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Quinasas S6 Ribosómicas 90-kDa/inmunología , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Activación Transcripcional/efectos de los fármacos , Regulación hacia Arriba , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología
14.
Immunity ; 42(6): 1100-15, 2015 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-26084025

RESUMEN

Tertiary lymphoid organs (TLOs) emerge during nonresolving peripheral inflammation, but their impact on disease progression remains unknown. We have found in aged Apoe(-/-) mice that artery TLOs (ATLOs) controlled highly territorialized aorta T cell responses. ATLOs promoted T cell recruitment, primed CD4(+) T cells, generated CD4(+), CD8(+), T regulatory (Treg) effector and central memory cells, converted naive CD4(+) T cells into induced Treg cells, and presented antigen by an unusual set of dendritic cells and B cells. Meanwhile, vascular smooth muscle cell lymphotoxin ß receptors (VSMC-LTßRs) protected against atherosclerosis by maintaining structure, cellularity, and size of ATLOs though VSMC-LTßRs did not affect secondary lymphoid organs: Atherosclerosis was markedly exacerbated in Apoe(-/-)Ltbr(-/-) and to a similar extent in aged Apoe(-/-)Ltbr(fl/fl)Tagln-cre mice. These data support the conclusion that the immune system employs ATLOs to organize aorta T cell homeostasis during aging and that VSMC-LTßRs participate in atherosclerosis protection via ATLOs.


Asunto(s)
Envejecimiento/inmunología , Aterosclerosis/inmunología , Receptor beta de Linfotoxina/metabolismo , Miocitos del Músculo Liso/fisiología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Adventicia/inmunología , Envejecimiento/genética , Animales , Aorta/patología , Apolipoproteínas E/genética , Aterosclerosis/genética , Diferenciación Celular/genética , Movimiento Celular/genética , Células Cultivadas , Coristoma/inmunología , Memoria Inmunológica , Activación de Linfocitos/genética , Tejido Linfoide/inmunología , Receptor beta de Linfotoxina/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas de Microfilamentos/genética , Proteínas Musculares/genética
15.
Immunol Cell Biol ; 93(6): 558-66, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25601276

RESUMEN

The cross talk between thymocytes and the thymic epithelium is critical for T-cell development and the establishment of central tolerance. Medullary thymic epithelial cells (mTECs) are located in the thymic medulla and mediate the elimination of self-reactive thymocytes, thereby preventing the onset of autoimmunity. Previous studies identified the deubiquitinating enzyme CYLD as a critical regulator of T-cell development by activating proximal T-cell receptor signaling during the transition of double-positive to single-positive thymocytes. Here we evaluated the impact of the naturally occurring short-splice variant of the cyld gene (sCYLD) on the development and maturation of mTECs. We found that thymi of CYLD(ex7/8) mice, solely expressing sCYLD, displayed a reduced number of mature mTECs caused by a developmental block during the transition of immature to mature mTECs. Further, we could demonstrate an impaired negative selection of thymocytes in these mice. Our data demonstrate that inefficient negative selection in the thymus of CYLD(ex7/8) mice result from a defect in mTEC maturation.


Asunto(s)
Diferenciación Celular , Cisteína Endopeptidasas/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Timo/citología , Timo/metabolismo , Animales , Antígenos de Superficie/metabolismo , Recuento de Células , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Cisteína Endopeptidasas/genética , Enzima Desubiquitinante CYLD , Femenino , Inmunofenotipificación , Ratones , Ratones Noqueados , Mutación , Fenotipo , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Timocitos/inmunología , Timocitos/metabolismo , Ubiquitinación
17.
Cell Rep ; 9(5): 1756-1769, 2014 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-25482563

RESUMEN

The mechanisms that govern proteolytic maturation or complete destruction of the precursor proteins p100 and p105 are fundamental to homeostasis and activation of NF-κB; however, they remain poorly understood. Using mass-spectrometry-based quantitative analysis of noncanonical LTßR-induced signaling, we demonstrate that stimulation induces simultaneous processing of both p100 and p105. The precursors not only form hetero-oligomers but also interact with the ATPase VCP/p97, and their induced proteolysis strictly depends on the signal response domain (SRD) of p100, suggesting that the SRD-targeting proteolytic machinery acts in cis and in trans. Separation of cellular pools by isotope labeling revealed synchronous dynamics of p105 and p100 proteolysis. The generation of p50 and p52 from their precursors depends on functional VCP/p97. We have developed quantitative mathematical models that describe the dynamics of the system and predict that p100-p105 complexes are signal responsive.


Asunto(s)
Subunidad p50 de NF-kappa B/metabolismo , Subunidad p52 de NF-kappa B/metabolismo , Precursores de Proteínas/metabolismo , Procesamiento Proteico-Postraduccional , Adenosina Trifosfatasas/metabolismo , Animales , Proteínas de Ciclo Celular/metabolismo , Células HEK293 , Células HeLa , Humanos , Cinética , Receptor beta de Linfotoxina/metabolismo , Ratones , Modelos Biológicos , Fosforilación , Proteolisis , Proteína que Contiene Valosina
18.
Neural Regen Res ; 9(7): 707-11, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25206877

RESUMEN

Activation of nuclear factor kappa B (NF-κB) is a hallmark of various central nervous system (CNS) pathologies. Neuron-specific inhibition of its transcriptional activator subunit RelA, also referred to as p65, promotes neuronal survival under a range of conditions, i.e., for ischemic or excitotoxic insults. In macro- and microglial cells, post-lesional activation of NF-κB triggers a growth-permissive program which contributes to neural tissue inflammation, scar formation, and the expression of axonal growth inhibitors. Intriguingly, inhibition of such inducible NF-κB in the neuro-glial compartment, i.e., by genetic ablation of RelA or overexpression of a transdominant negative mutant of its upstream regulator IκBα, significantly enhances functional recovery and promotes axonal regeneration in the mature CNS. By contrast, depletion of the NF-κB subunit p50, which lacks transcriptional activator function and acts as a transcriptional repressor on its own, causes precocious neuronal loss and exacerbates axonal degeneration in the lesioned brain. Collectively, the data imply that NF-κB orchestrates a multicellular program in which κB-dependent gene expression establishes a growth-repulsive terrain within the post-lesioned brain that limits structural regeneration of neuronal circuits. Considering these subunit-specific functions, interference with the NF-κB pathway might hold clinical potentials to improve functional restoration following traumatic CNS injury.

19.
J Vis Exp ; (89)2014 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-25080017

RESUMEN

The rodent visual system encompasses retinal ganglion cells and their axons that form the optic nerve to enter thalamic and midbrain centers, and postsynaptic projections to the visual cortex. Based on its distinct anatomical structure and convenient accessibility, it has become the favored structure for studies on neuronal survival, axonal regeneration, and synaptic plasticity. Recent advancements in MR imaging have enabled the in vivo visualization of the retino-tectal part of this projection using manganese mediated contrast enhancement (MEMRI). Here, we present a MEMRI protocol for illustration of the visual projection in mice, by which resolutions of (200 µm)3 can be achieved using common 3 Tesla scanners. We demonstrate how intravitreal injection of a single dosage of 15 nmol MnCl2 leads to a saturated enhancement of the intact projection within 24 hr. With exception of the retina, changes in signal intensity are independent of coincided visual stimulation or physiological aging. We further apply this technique to longitudinally monitor axonal degeneration in response to acute optic nerve injury, a paradigm by which Mn2+ transport completely arrests at the lesion site. Conversely, active Mn2+ transport is quantitatively proportionate to the viability, number, and electrical activity of axon fibers. For such an analysis, we exemplify Mn2+ transport kinetics along the visual path in a transgenic mouse model (NF-κB p50KO) displaying spontaneous atrophy of sensory, including visual, projections. In these mice, MEMRI indicates reduced but not delayed Mn2+ transport as compared to wild type mice, thus revealing signs of structural and/or functional impairments by NF-κB mutations. In summary, MEMRI conveniently bridges in vivo assays and post mortem histology for the characterization of nerve fiber integrity and activity. It is highly useful for longitudinal studies on axonal degeneration and regeneration, and investigations of mutant mice for genuine or inducible phenotypes.


Asunto(s)
Imagen por Resonancia Magnética/métodos , Nervio Óptico/anatomía & histología , Animales , Axones/fisiología , Medios de Contraste , Imagen por Resonancia Magnética/instrumentación , Ratones , Ratones Noqueados , Degeneración Nerviosa/patología , Nervio Óptico/citología , Enfermedades del Nervio Óptico/patología , Traumatismos del Nervio Óptico/patología , Células Ganglionares de la Retina/citología , Vías Visuales/anatomía & histología , Vías Visuales/citología
20.
J Cell Sci ; 127(Pt 14): 3052-65, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24860143

RESUMEN

NF-κB is dually involved in neurogenesis and brain pathology. Here, we addressed its role in adult axoneogenesis by generating mutations of RelA (p65) and p50 (also known as NFKB1) heterodimers of canonical NF-κB. In addition to RelA activation in astrocytes, optic nerve axonotmesis caused a hitherto unrecognized induction of RelA in growth-inhibitory oligodendrocytes. Intraretinally, RelA was induced in severed retinal ganglion cells and was also expressed in bystander Müller glia. Cell-type-specific deletion of transactivating RelA in neurons and/or macroglia stimulated axonal regeneration in a distinct and synergistic pattern. By contrast, deletion of the p50 suppressor subunit promoted spontaneous and post-injury Wallerian degeneration. Growth effects mediated by RelA deletion paralleled a downregulation of growth-inhibitory Cdh1 (officially known as FZR1) and upregulation of the endogenous Cdh1 suppressor EMI1 (officially known as FBXO5). Pro-degenerative loss of p50, however, stabilized retinal Cdh1. In vitro, RelA deletion elicited opposing pro-regenerative shifts in active nuclear and inactive cytoplasmic moieties of Cdh1 and Id2. The involvement of NF-κB and cell-cycle regulators such as Cdh1 in regenerative processes of non-replicative neurons suggests novel mechanisms by which molecular reprogramming might be executed to stimulate adult axoneogenesis and treat central nervous system (CNS) axonopathies.


Asunto(s)
Axones/fisiología , Sistema Nervioso Central/metabolismo , Subunidad p50 de NF-kappa B/metabolismo , Regeneración Nerviosa/fisiología , Células Ganglionares de la Retina/fisiología , Factor de Transcripción ReIA/metabolismo , Animales , Transporte Axonal , Axones/metabolismo , Proteínas Cdh1/metabolismo , Proteína 2 Inhibidora de la Diferenciación/metabolismo , Ratones , Ratones Transgénicos , Células Ganglionares de la Retina/metabolismo , Degeneración Walleriana/metabolismo , Degeneración Walleriana/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...